S. Lista, N. Toschi, F. Baldacci, H. Zetterberg, K. Blennow et al., Cerebrospinal Fluid Neurogranin as a Biomarker of Neurodegenerative Diseases: A Cross-Sectional Study, J Alzheimers Dis, vol.59, pp.1327-1334, 2017.

-. Spires, L. Tara, H. Bradley, and T. , The Intersection of Amyloid Beta and Tau at Synapses in Alzheimer's Disease, Neuron, vol.82, pp.756-771, 2014.

M. Husain, Alzheimer's disease: time to focus on the brain, not just molecules, Brain, vol.140, pp.251-253, 2017.

M. J. Legato, P. A. Johnson, and J. E. Manson, Consideration of Sex Differences in Medicine to Improve Health Care and Patient Outcomes, JAMA, vol.316, p.1865, 2016.

K. Irvine, K. R. Laws, T. M. Gale, and T. K. Kondel, Greater cognitive deterioration in women than men with Alzheimer's disease: A meta analysis, J Clin Exp Neuropsychol, vol.34, pp.989-998, 2012.

K. A. Lin, K. R. Choudhury, B. G. Rathakrishnan, D. M. Marks, J. R. Petrella et al.,

, Alzheimer's Disease Neuroimaging I. Marked gender differences in progression of mild cognitive impairment over 8 years, Alzheimers Dement (N Y), vol.1, pp.103-110, 2015.

X. Hua, D. P. Hibar, S. Lee, A. W. Toga, C. R. Jack et al., Sex and age differences in atrophic rates: an ADNI study with n=1368 MRI scans, Neurobiol Aging, vol.31, pp.1463-1480, 2010.

N. Mattsson, A. Lönneborg, M. Boccardi, K. Blennow, and O. Hansson, Clinical validity of cerebrospinal fluid A?42, tau, and phospho-tau as biomarkers for Alzheimer's disease in the context of a structured 5-phase development framework, Neurobiol Aging, vol.52, pp.196-213, 2017.

A. Altmann, L. Tian, V. W. Henderson, and M. D. Greicius, Sex modifies theAPOE-related risk of developing Alzheimer disease, Ann Neurol, vol.75, pp.563-573, 2014.

H. Depypere, A. Vierin, S. Weyers, and A. Sieben, Alzheimer's disease, apolipoprotein E and hormone replacement therapy, Maturitas, vol.94, pp.98-105, 2016.

R. Scacchi, G. Gambina, E. Broggio, and R. M. Corbo, Sex and ESR1 genotype may influence the response to treatment with donepezil and rivastigmine in patients with Alzheimer's disease, Int J Geriatr Psychiatry, vol.29, pp.610-615, 2013.

, Alzheimer's disease facts and figures, Alzheimer's & Dementia, vol.10, pp.47-92, 2014.

H. Hampel, O. Bryant, S. E. Castrillo, J. I. Ritchie, C. Rojkova et al., PRECISION MEDICINE-The Golden Gate for Detection, Treatment and Prevention of Alzheimer's Disease, J Prev Alzheimers Dis, vol.3, pp.243-259, 2016.

H. Hampel, O. Bryant, S. E. Durrleman, S. Younesi, E. Rojkova et al., A Precision Medicine Initiative for Alzheimer's disease: the road ahead to biomarker-guided integrative disease modeling, Climacteric, vol.20, pp.107-118, 2017.

S. Lista, Z. S. Khachaturian, D. Rujescu, F. Garaci, B. Dubois et al., Application of Systems Theory in Longitudinal Studies on the Origin and Progression of Alzheimer's Disease, Methods Mol Biol, vol.1303, pp.49-67, 2016.

E. Younesi and M. Hofmann-apitius, From integrative disease modeling to predictive, preventive, personalized and participatory (P4) medicine, EPMA Journal, issue.4, 2013.

, FDA approves first cancer treatment for any solid tumor with a specific genetic feature, 2017.

K. Imamura, Y. Izumi, A. Watanabe, K. Tsukita, K. Woltjen et al., The Src/c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis, Sci Transl Med, vol.9, p.3962, 2017.

, certain-cancer-drugs-held-out-prospects-als-therapy variables for blood-based biomarker studies in Alzheimer's disease research, Certain Cancer Drugs Held Out as Prospects for ALS Therapy, vol.11, pp.549-560, 2015.

K. Henriksen, O. Bryant, S. E. Hampel, H. Trojanowski, J. Q. Montine et al., The future of blood-based biomarkers for Alzheimer's disease, Alzheimer's & Dementia, vol.10, pp.115-131, 2014.

H. M. Snyder, M. C. Carrillo, F. Grodstein, K. Henriksen, A. Jeromin et al., Developing novel blood-based biomarkers for Alzheimer's disease, Alzheimer's & Dementia, vol.10, pp.109-114, 2014.

V. Gupta, R. Sundaram, and R. N. Martins, Multiplex biomarkers in blood, Alzheimers Res Ther, vol.5, p.31, 2013.

S. Lista, F. Faltraco, D. Prvulovic, and H. Hampel, Blood and plasma-based proteomic biomarker research in Alzheimer's disease, Prog Neurobiol, pp.101-102, 2013.

S. Lista, F. Faltraco, and H. Hampel, Biological and methodical challenges of blood-based proteomics in the field of neurological research, Prog Neurobiol, pp.18-34, 2013.

S. J. Teipel, M. Grothe, S. Lista, N. Toschi, F. G. Garaci et al., Relevance of Magnetic Resonance Imaging for Early Detection and Diagnosis of Alzheimer Disease, Med Clin North Am, vol.97, pp.399-424, 2013.

M. Ewers, R. A. Sperling, W. E. Klunk, M. W. Weiner, and H. Hampel, Neuroimaging markers for the prediction and early diagnosis of Alzheimer's disease dementia, Trends Neurosci, vol.34, pp.430-442, 2011.

M. Ewers, G. B. Frisoni, S. J. Teipel, L. T. Grinberg, E. Amaro et al., Staging Alzheimer's disease progression with multimodality neuroimaging, Prog Neurobiol, vol.95, pp.535-546, 2011.

H. Hampel, D. Prvulovic, S. J. Teipel, and A. Bokde, Recent developments of functional magnetic resonance imaging research for drug development in Alzheimer's disease, Prog Neurobiol, vol.95, pp.570-578, 2011.

P. A. Chiesa, E. Cavedo, S. Lista, P. M. Thompson, H. Hampel et al., Revolution of RestingState Functional Neuroimaging Genetics in Alzheimer's Disease, Trends Neurosci, vol.40, pp.469-480, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01560182

M. Engels, W. M. Van-der-flier, C. J. Stam, A. Hillebrand, P. Scheltens et al., Alzheimer's disease: The state of the art in resting-state magnetoencephalography, Clin Neurophysiol, vol.128, pp.1426-1437, 2017.

K. Broich, M. Weiergräber, and H. Hampel, Biomarkers in clinical trials for neurodegenerative diseases: Regulatory perspectives and requirements, Prog Neurobiol, vol.95, pp.498-500, 2011.

G. Nistico, K. Broich, and H. Hampel, Need for new guidelines for Alzheimer's disease clinical trials, Eur J Neurodegener Dis, vol.2, pp.181-186, 2013.

J. T. Jørgensen, Companion diagnostics: the key to personalized medicine, Expert Review of Molecular Diagnostics, vol.15, pp.153-156, 2015.

G. H. Lyman and H. L. Moses, Biomarker Tests for Molecularly Targeted Therapies-The Key to Unlocking Precision Medicine, N Engl J Med, vol.375, pp.4-6, 2016.

A. K. Daly, Pharmacogenetics and human genetic polymorphisms, Biochem J, vol.429, pp.435-449, 2010.

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.

V. Escott-price, M. Shoai, R. Pither, J. Williams, and J. Hardy, Polygenic score prediction captures nearly all common genetic risk for Alzheimer's disease, Neurobiol Aging, vol.49, pp.214-217, 2017.

H. L. Rehm, Evolving health care through personal genomics, Nature Reviews Genetics, vol.18, pp.259-267, 2017.

O. Sporns, Networks of the Brain, 2010.

D. C. Van-essen, S. M. Smith, D. M. Barch, T. E. Behrens, Y. E. Ugurbil et al., The WU-Minn human connectome project: an overview, Neuroimage, vol.80, pp.62-79, 2013.

X. Zuo, Y. He, R. F. Betzel, S. Colcombe, O. Sporns et al., Human connectomics across the life span, Trends in cognitive sciences, vol.21, pp.32-45, 2017.

A. Fornito, A. Zalesky, and E. Bullmore, Fundamentals of brain network analysis, 2016.

L. Pini, M. Pievani, M. Bocchetta, D. Altomare, P. Bosco et al., Brain atrophy in Alzheimer's disease and aging, Ageing research reviews, vol.30, pp.25-48, 2016.

E. Cavedo and G. Frisoni, The dynamic marker hypothesis of Alzheimer's disease and its implications for clinical imaging. The quarterly journal of nuclear medicine and molecular imaging: official publication of the Italian Association of Nuclear Medicine (AIMN) [and] the International Association of Radiopharmacology (IAR), [and] Section of the Society of, vol.55, pp.237-249, 2011.

E. Cavedo, A. Redolfi, F. Angeloni, C. Babiloni, R. Lizio et al., The Italian Alzheimer's Disease Neuroimaging Initiative (I-ADNI): validation of structural MR imaging, J Alzheimers Dis, vol.40, pp.941-952, 2014.

C. R. Jack, M. A. Bernstein, B. J. Borowski, J. L. Gunter, N. C. Fox et al., Update on the magnetic resonance imaging core of the Alzheimer's disease neuroimaging initiative, Alzheimer's & Dementia, vol.6, pp.212-220, 2010.

H. Braak and E. Braak, Staging of alzheimer's disease-related neurofibrillary changes, Neurobiol Aging, vol.16, pp.271-278, 1995.

A. Delacourte, J. P. David, N. Sergeant, L. Buee, A. Wattez et al., The biochemical pathway of neurofibrillary degeneration in aging and Alzheimer's disease, Neurology, vol.52, pp.1158-1158, 1999.

L. C. Souza, M. Chupin, F. Lamari, C. Jardel, D. Leclercq et al., CSF tau markers are correlated with hippocampal volume in Alzheimer's disease, Neurobiol Aging, vol.33, pp.1253-1257, 2012.

C. R. Jack, V. J. Lowe, M. L. Senjem, S. D. Weigand, B. J. Kemp et al., 11C PiB and structural MRI provide complementary information in imaging of Alzheimer's disease and amnestic mild cognitive impairment, Brain, vol.131, pp.665-680, 2008.

K. A. Josephs, J. L. Whitwell, Z. Ahmed, M. M. Shiung, S. D. Weigand et al., ?-amyloid burden is not associated with rates of brain atrophy, Ann Neurol, vol.63, pp.204-212, 2008.

L. Ferrarini, W. M. Palm, H. Olofsen, R. Van-der-landen, J. Blauw et al., Admiraal-Behloul F. MMSE scores correlate with local ventricular enlargement in the spectrum from cognitively normal to Alzheimer disease, Neuroimage, vol.39, pp.1832-1838, 2008.

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, vol.22, pp.285-294, 2010.

N. C. Fox, S. Cousens, R. Scahill, R. J. Harvey, and M. N. Rossor, Using Serial Registered Brain Magnetic Resonance Imaging to Measure Disease Progression in Alzheimer Disease, Arch Neurol, vol.57, p.339, 2000.

N. C. Fox, R. I. Scahill, W. R. Crum, and M. N. Rossor, Correlation between rates of brain atrophy and cognitive decline in AD, Neurology, vol.52, pp.1687-1687, 1999.

C. R. Jack, R. C. Petersen, M. Grundman, J. S. Gamst, A. Ward et al., Longitudinal MRI findings from the vitamin E and donepezil treatment study for MCI, Neurobiol Aging, vol.29, pp.1285-1295, 2008.

J. C. Baron, G. Chételat, B. Desgranges, G. Perchey, B. Landeau et al., In Vivo Mapping of Gray Matter Loss with Voxel-Based Morphometry in Mild Alzheimer's Disease, Neuroimage, vol.14, pp.298-309, 2001.

V. Singh, H. Chertkow, J. P. Lerch, A. C. Evans, A. E. Dorr et al., Spatial patterns of cortical thinning in mild cognitive impairment and Alzheimer's disease, Brain, vol.129, pp.2885-2893, 2006.

R. Cuingnet, J. A. Glaunes, M. Chupin, H. Benali, and O. Colliot, Spatial and Anatomical Regularization of SVM: A General Framework for Neuroimaging Data, IEEE Transactions on Pattern Analysis and Machine Intelligence, vol.35, pp.682-696, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00790079

C. Davatzikos, Y. Fan, X. Wu, D. Shen, and S. M. Resnick, Detection of prodromal Alzheimer's disease via pattern classification of magnetic resonance imaging, Neurobiol Aging, vol.29, pp.514-523, 2008.

S. Kloppel, C. M. Stonnington, C. Chu, B. Draganski, R. I. Scahill et al., Automatic classification of MR scans in Alzheimer's disease, Brain, vol.131, pp.681-689, 2008.

C. Plant, S. J. Teipel, A. Oswald, C. Böhm, T. Meindl et al., Automated detection of brain atrophy patterns based on MRI for the prediction of Alzheimer's disease, Neuroimage, vol.50, pp.162-174, 2010.

E. Cavedo, M. Boccardi, R. Ganzola, E. Canu, A. Beltramello et al., Local amygdala structural differences with 3T MRI in patients with Alzheimer disease, Neurology, vol.76, pp.727-733, 2011.

G. B. Frisoni, C. R. Jack, M. Bocchetta, C. Bauer, K. S. Frederiksen et al., The EADC-ADNI Harmonized Protocol for manual hippocampal segmentation on magnetic resonance: evidence of validity, Alzheimer's & Dementia, vol.11, pp.111-125, 2015.

P. Scheltens, L. J. Launer, F. Barkhof, H. C. Weinstein, and W. A. Gool, Visual assessment of medial temporal lobe atrophy on magnetic resonance imaging: Interobserver reliability, J Neurol, vol.242, pp.557-560, 1995.

E. Cavedo, P. Suppa, C. Lange, R. Opfer, S. Lista et al., Fully Automatic MRI-Based Hippocampus Volumetry Using FSL-FIRST: IntraScanner Test-Retest Stability, Inter-Field Strength Variability, and Performance as Enrichment Biomarker for Clinical Trials Using Prodromal Target Populations at Risk for Alzheimer's Disease, J Alzheimers Dis, vol.60, pp.151-164, 2017.

K. Ritter, C. Lange, M. Weygandt, A. Mäurer, A. Roberts et al., Combination of Structural MRI and FDG-PET of the Brain Improves Diagnostic Accuracy in Newly Manifested Cognitive Impairment in Geriatric Inpatients, J Alzheimers Dis, vol.54, pp.1319-1331, 2016.

P. Suppa, H. Hampel, T. Kepp, C. Lange, L. Spies et al., Performance of hippocampus volumetry with FSL-first for prediction of Alzheimer's disease dementia in at risk subjects with amnestic mild cognitive impairment, J Alzheimers Dis, vol.51, pp.867-873, 2016.

P. Suppa, U. Anker, L. Spies, I. Bopp, B. Rüegger-frey et al., Fully automated atlas-based hippocampal volumetry for detection of Alzheimer's disease in a memory clinic setting, J Alzheimers Dis, vol.44, pp.183-193, 2015.

C. Boutet, M. Chupin, O. Colliot, M. Sarazin, G. Mutlu et al., Is radiological evaluation as good as computer-based volumetry to assess hippocampal atrophy in Alzheimer's disease?, Neuroradiology, vol.54, pp.1321-1330, 2012.

O. Colliot, G. Chételat, M. Chupin, B. Desgranges, B. Magnin et al., Discrimination between Alzheimer Disease, Mild Cognitive Impairment, and Normal Aging by Using Automated Segmentation of the Hippocampus, Radiology, vol.248, pp.194-201, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00383265

A. Bakkour, J. C. Morris, and B. C. Dickerson, The cortical signature of prodromal AD: Regional thinning predicts mild AD dementia, Neurology, vol.72, pp.1048-1055, 2008.

R. S. Desikan, H. J. Cabral, C. P. Hess, W. P. Dillon, C. M. Glastonbury et al., Automated MRI measures identify individuals with mild cognitive impairment and Alzheimer's disease, Brain, vol.132, pp.2048-2057, 2009.

C. R. Jack, R. C. Petersen, Y. C. Xu, P. C. O'brien, G. E. Smith et al., Prediction of AD with MRI-based hippocampal volume in mild cognitive impairment, Neurology, vol.52, pp.1397-1397, 1999.

S. M. Landau, D. Harvey, C. M. Madison, E. M. Reiman, N. L. Foster et al., Comparing predictors of conversion and decline in mild cognitive impairment, Neurology, vol.75, pp.230-238, 2010.

P. Visser, F. Verhey, P. Hofman, P. Scheltens, and J. Jolles, Medial temporal lobe atrophy predicts Alzheimer9s disease in patients with minor cognitive impairment, J Neurol Neurosurg Psychiatry, vol.72, pp.491-497, 2002.

L. De-souza, M. Chupin, M. Bertoux, S. Lehéricy, B. Dubois et al., Is hippocampal volume a good marker to differentiate Alzheimer's disease from frontotemporal dementia?, Alzheimer's & Dementia, vol.9, pp.380-381, 2013.

M. Hashimoto, H. Kitagaki, T. Imamura, N. Hirono, T. Shimomura et al., Medial temporal and whole-brain atrophy in dementia with Lewy bodies: A volumetric MRI study, Neurology, vol.51, pp.357-362, 1998.

C. Boutet, M. Chupin, S. Lehéricy, L. Marrakchi-kacem, S. Epelbaum et al., Detection of volume loss in hippocampal layers in Alzheimer's disease using 7 T MRI: A feasibility study, NeuroImage: Clinical, vol.5, pp.341-348, 2014.

L. Wisse, G. J. Biessels, S. M. Heringa, H. J. Kuijf, D. L. Koek et al., Hippocampal subfield volumes at 7T in early Alzheimer's disease and normal aging, Neurobiol Aging, vol.35, pp.2039-2045, 2014.

I. Kilimann, M. Grothe, H. Heinsen, E. Alho, L. Grinberg et al., Subregional basal forebrain atrophy in Alzheimer's disease: a multicenter study, J Alzheimers Dis, vol.40, pp.687-700, 2014.

H. Hampel, M. M. Mesulam, A. C. Cuello, A. S. Khachaturian, M. R. Farlow et al., Revisiting the cholinergic hypothesis in Alzheimer's disease: Emerging evidence from translational and clinical research, 2017.

K. Brüggen, M. Dyrba, F. Barkhof, L. Hausner, M. Filippi et al., Basal forebrain and hippocampus as predictors of conversion to Alzheimer's disease in patients with mild cognitive impairment-A multicenter DTI and volumetry study, J Alzheimers Dis, vol.48, pp.197-204, 2015.

M. Grothe, H. Heinsen, and S. J. Teipel, Atrophy of the cholinergic basal forebrain over the adult age range and in early stages of Alzheimer's disease, Biol Psychiatry, vol.71, pp.805-813, 2012.

H. Zhang, J. N. Trollor, W. Wen, W. Zhu, J. D. Crawford et al., Grey matter atrophy of basal forebrain and hippocampus in mild cognitive impairment, J Neurol Neurosurg Psychiatry, vol.82, pp.487-493, 2011.

M. J. Grothe, H. Heinsen, E. Amaro, L. T. Grinberg, and S. J. Teipel, Cognitive correlates of basal forebrain atrophy and associated cortical hypometabolism in mild cognitive impairment, Cereb Cortex, vol.26, pp.2411-2426, 2015.

S. J. Teipel, T. Meindl, L. Grinberg, M. Grothe, J. L. Cantero et al., The cholinergic system in mild cognitive impairment and Alzheimer's disease: an in vivo MRI and DTI study, Hum Brain Mapp, vol.32, pp.1349-1362, 2011.

S. J. Teipel, W. H. Flatz, H. Heinsen, A. L. Bokde, S. O. Schoenberg et al., Measurement of basal forebrain atrophy in Alzheimer's disease using MRI, Brain, vol.128, pp.2626-2644, 2005.

T. W. Schmitz and R. N. Spreng, Initiative AsDN. Basal forebrain degeneration precedes and predicts the cortical spread of Alzheimer's pathology, Nature communications, p.7, 2016.

M. J. Grothe, M. Ewers, B. Krause, H. Heinsen, and S. J. Teipel, Initiative AsDN. Basal forebrain atrophy and cortical amyloid deposition in nondemented elderly subjects, Alzheimer's & Dementia, vol.10, pp.344-353, 2014.

G. M. Kerbler, J. Fripp, C. C. Rowe, V. L. Villemagne, O. Salvado et al., Initiative AsDN. Basal forebrain atrophy correlates with amyloid ? burden in Alzheimer's disease, NeuroImage: Clinical, vol.7, pp.105-113, 2015.

R. Cuingnet, E. Gerardin, J. Tessieras, G. Auzias, S. Lehéricy et al., Automatic classification of patients with Alzheimer's disease from structural MRI: A comparison of ten methods using the ADNI database, Neuroimage, vol.56, pp.766-781, 2011.

C. Davatzikos, P. Bhatt, L. M. Shaw, K. N. Batmanghelich, and J. Q. Trojanowski, Prediction of MCI to AD conversion, via MRI, CSF biomarkers, and pattern classification, Neurobiol Aging, vol.32, pp.2322-2319, 2011.

Z. Sun, M. Van-de-giessen, B. Lelieveldt, and M. Staring, Detection of Conversion from Mild Cognitive Impairment to Alzheimer's Disease Using Longitudinal Brain MRI. Front Neuroinform, p.11, 2017.

E. Cavedo, B. Dubois, O. Colliot, S. Lista, B. Croisile et al., Reduced Regional Cortical Thickness Rate of Change in Donepezil-Treated Subjects With Suspected Prodromal Alzheimer's Disease, The Journal of Clinical Psychiatry, vol.77, pp.1631-1638, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01388951

B. Dubois, M. Chupin, H. Hampel, S. Lista, E. Cavedo et al., Donepezil decreases annual rate of hippocampal atrophy in suspected prodromal Alzheimer's disease, Alzheimer's & Dementia, vol.11, pp.1041-1049, 2015.

E. Cavedo, M. J. Grothe, O. Colliot, S. Lista, M. Chupin et al., Reduced basal forebrain atrophy progression in a randomized Donepezil trial in prodromal Alzheimer's disease, Sci Rep, vol.7, p.11706, 2017.

N. C. Fox, R. S. Black, S. Gilman, M. N. Rossor, S. G. Griffith et al., Effects of A immunization (AN1792) on MRI measures of cerebral volume in Alzheimer disease, Neurology, vol.64, pp.1563-1572, 2005.

R. Sperling, S. Salloway, D. J. Brooks, D. Tampieri, J. Barakos et al., Amyloid-related imaging abnormalities in patients with Alzheimer's disease treated with bapineuzumab: a retrospective analysis, The Lancet Neurology, vol.11, pp.241-249, 2012.

M. Bozzali and A. Cherubini, Diffusion tensor MRI to investigate dementias: a brief review, Magn Reson Imaging, vol.25, pp.969-977, 2007.

T. C. Chua, W. Wen, M. J. Slavin, and P. S. Sachdev, Diffusion tensor imaging in mild cognitive impairment and Alzheimer's disease: a review, Curr Opin Neurol, vol.21, pp.83-92, 2008.

C. P. Hess, Update on diffusion tensor imaging in Alzheimer's disease, Magn Reson Imaging Clin N Am, vol.17, pp.215-224, 2009.

P. G. Nucifora, R. Verma, S. K. Lee, and E. R. Melhem, Diffusion-tensor MR imaging and tractography: exploring brain microstructure and connectivity, Radiology, vol.245, pp.367-384, 2007.
DOI : 10.1148/radiol.2452060445

F. G. Garaci, F. Bolacchi, A. Cerulli, M. Melis, A. Spano et al., Optic nerve and optic radiation neurodegeneration in patients with glaucoma: in vivo analysis with 3-T diffusion-tensor MR imaging, Radiology, vol.252, pp.496-501, 2009.

M. Pardini, M. Elia, F. G. Garaci, S. Guida, F. Coniglione et al., Long-term cognitive and behavioral therapies, combined with augmentative communication, are related to uncinate fasciculus integrity in autism, J Autism Dev Disord, vol.42, pp.585-592, 2012.

M. Pardini, F. G. Garaci, L. Bonzano, L. Roccatagliata, M. G. Palmieri et al., White matter reduced streamline coherence in young men with autism and mental retardation, Eur J Neurol, vol.16, pp.1185-1190, 2009.
DOI : 10.1111/j.1468-1331.2009.02699.x

D. K. Jones and A. Leemans, Diffusion tensor imaging, Methods Mol Biol, vol.711, pp.127-144, 2011.

D. K. Jones, Studying connections in the living human brain with diffusion MRI, Cortex, vol.44, pp.936-952, 2008.

B. Jeurissen, J. D. Tournier, T. Dhollander, A. Connelly, and J. Sijbers, Multi-tissue constrained spherical deconvolution for improved analysis of multi-shell diffusion MRI data, Neuroimage, vol.103, pp.411-426, 2014.

R. E. Smith, J. D. Tournier, F. Calamante, and A. Connelly, SIFT: Spherical-deconvolution informed filtering of tractograms, Neuroimage, vol.67, pp.298-312, 2013.
DOI : 10.1016/j.neuroimage.2012.11.049

B. Jeurissen, A. Leemans, D. K. Jones, J. D. Tournier, and J. Sijbers, Probabilistic fiber tracking using the residual bootstrap with constrained spherical deconvolution, Hum Brain Mapp, vol.32, pp.461-479, 2011.
DOI : 10.1002/hbm.21032

J. D. Tournier, C. H. Yeh, F. Calamante, K. H. Cho, A. Connelly et al., Resolving crossing fibres using constrained spherical deconvolution: validation using diffusion-weighted imaging phantom data, Neuroimage, vol.42, pp.617-625, 2008.
DOI : 10.1016/j.neuroimage.2008.05.002

M. Giannelli, G. Belmonte, N. Toschi, I. Pesaresi, P. Ghedin et al., Technical note: DTI measurements of fractional anisotropy and mean diffusivity at 1.5 T: comparison of two radiofrequency head coils with different functional designs and sensitivities, Med Phys, vol.38, pp.3205-3211, 2011.

D. Medina, L. Detoledo-morrell, F. Urresta, J. D. Gabrieli, M. Moseley et al., White matter changes in mild cognitive impairment and AD: A diffusion tensor imaging study, Neurobiol Aging, vol.27, pp.663-672, 2006.
DOI : 10.1016/j.neurobiolaging.2005.03.026

Y. Liu, G. Spulber, K. K. Lehtimaki, M. Kononen, I. Hallikainen et al., Diffusion tensor imaging and tract-based spatial statistics in Alzheimer's disease and mild cognitive impairment, Neurobiol Aging, vol.32, pp.1558-1571, 2011.
DOI : 10.1016/j.neurobiolaging.2009.10.006

A. M. Fjell, I. K. Amlien, L. T. Westlye, and K. B. Walhovd, Mini-mental state examination is sensitive to brain atrophy in Alzheimer's disease, Dement Geriatr Cogn Disord, vol.28, pp.252-258, 2009.

J. H. Heo, S. T. Lee, C. Kon, H. J. Park, J. Y. Shim et al., White matter hyperintensities and cognitive dysfunction in Alzheimer disease, J Geriatr Psychiatry Neurol, vol.22, pp.207-212, 2009.
DOI : 10.1177/0891988709335800

T. C. Chua, W. Wen, X. Chen, N. Kochan, M. J. Slavin et al., Diffusion tensor imaging of the posterior cingulate is a useful biomarker of mild cognitive impairment, Am J Geriatr Psychiatry, vol.17, pp.602-613, 2009.

S. Takahashi, H. Yonezawa, J. Takahashi, M. Kudo, T. Inoue et al., Selective reduction of diffusion anisotropy in white matter of Alzheimer disease brains measured by 3.0 Tesla magnetic resonance imaging, Neurosci Lett, vol.332, pp.45-48, 2002.

Y. Zhang, N. Schuff, G. H. Jahng, W. Bayne, S. Mori et al., Diffusion tensor imaging of cingulum fibers in mild cognitive impairment and Alzheimer disease, Neurology, vol.68, pp.13-19, 2007.

L. Zhuang, W. Wen, W. Zhu, J. Trollor, N. Kochan et al., White matter integrity in mild cognitive impairment: a tract-based spatial statistics study, Neuroimage, vol.53, pp.16-25, 2010.
DOI : 10.1016/j.neuroimage.2010.05.068

G. Douaud, S. Jbabdi, T. E. Behrens, R. A. Menke, A. Gass et al., DTI measures in crossing-fibre areas: increased diffusion anisotropy reveals early white matter alteration in MCI and mild Alzheimer's disease, Neuroimage, vol.55, pp.880-890, 2011.
DOI : 10.1016/j.neuroimage.2010.12.008

D. B. Ennis and G. Kindlmann, Orthogonal tensor invariants and the analysis of diffusion tensor magnetic resonance images, Magn Reson Med, vol.55, pp.136-146, 2006.

N. H. Stricker, B. C. Schweinsburg, L. Delano-wood, C. E. Wierenga, K. J. Bangen et al., Decreased white matter integrity in late-myelinating fiber pathways in Alzheimer's disease supports retrogenesis, Neuroimage, vol.45, pp.10-16, 2009.
DOI : 10.1016/j.neuroimage.2008.11.027

URL : http://europepmc.org/articles/pmc2782417?pdf=render

S. J. Teipel, R. Stahl, O. Dietrich, S. O. Schoenberg, R. Perneczky et al., Multivariate network analysis of fiber tract integrity in Alzheimer's disease, Neuroimage, vol.34, pp.985-995, 2007.

P. Selnes, D. Aarsland, A. Bjornerud, L. Gjerstad, A. Wallin et al., Diffusion tensor imaging surpasses cerebrospinal fluid as predictor of cognitive decline and medial temporal lobe atrophy in subjective cognitive impairment and mild cognitive impairment, J Alzheimers Dis, vol.33, pp.723-736, 2013.

C. E. Sexton, U. G. Kalu, N. Filippini, C. E. Mackay, and K. P. Ebmeier, A meta-analysis of diffusion tensor imaging in mild cognitive impairment and Alzheimer's disease, Neurobiol Aging, vol.32, pp.2322-2325, 2011.

S. J. Teipel, S. Reuter, B. Stieltjes, J. Acosta-cabronero, U. Ernemann et al., Multicenter stability of diffusion tensor imaging measures: A European clinical and physical phantom study, Psychiatry Res, vol.194, pp.363-371, 2011.
DOI : 10.1016/j.pscychresns.2011.05.012

M. M. Mielke, O. C. Okonkwo, K. Oishi, S. Mori, S. Tighe et al., Fornix integrity and hippocampal volume predict memory decline and progression to Alzheimer's disease, Alzheimers Dement, vol.8, pp.105-113, 2012.
DOI : 10.1016/j.jalz.2011.05.2416

URL : http://europepmc.org/articles/pmc3305232?pdf=render

G. Douaud, R. A. Menke, A. Gass, A. U. Monsch, A. Rao et al., Brain Microstructure Reveals Early Abnormalities more than Two Years prior to Clinical Progression from Mild Cognitive Impairment to Alzheimer's Disease, J Neurosci, vol.33, pp.2147-2155, 2013.
DOI : 10.1523/jneurosci.4437-12.2013

URL : http://www.jneurosci.org/content/33/5/2147.full.pdf

J. L. Andersson, S. Skare, and J. Ashburner, How to correct susceptibility distortions in spin-echo echoplanar images: application to diffusion tensor imaging, Neuroimage, vol.20, pp.870-888, 2003.
DOI : 10.1016/s1053-8119(03)00336-7

C. Eichner, K. Setsompop, P. J. Koopmans, R. Lutzkendorf, D. G. Norris et al., Slice accelerated diffusion-weighted imaging at ultra-high field strength, Magn Reson Med, vol.71, pp.1518-1525, 2014.
DOI : 10.1002/mrm.24809

S. A. Back, C. D. Kroenke, L. S. Sherman, G. Lawrence, X. Gong et al., White matter lesions defined by diffusion tensor imaging in older adults, Ann Neurol, vol.70, pp.465-476, 2011.
DOI : 10.1002/ana.22484

URL : http://europepmc.org/articles/pmc3177155?pdf=render

M. Cosottini, D. Frosini, I. Pesaresi, M. Costagli, L. Biagi et al., MR Imaging of the Substantia Nigra at 7 T Enables Diagnosis of Parkinson Disease, Radiology, vol.271, pp.831-838, 2014.

E. Verstraete, D. L. Polders, R. C. Mandl, . Van-den, M. P. Heuvel et al., Multimodal tract-based analysis in ALS patients at 7T: a specific white matter profile?, Amyotroph Lateral Scler Frontotemporal Degener, vol.15, pp.84-92, 2014.

A. L. Alexander, K. M. Hasan, M. Lazar, J. S. Tsuruda, and D. L. Parker, Analysis of partial volume effects in diffusion-tensor MRI, Magn Reson Med, vol.45, pp.770-780, 2001.

V. J. Wedeen, R. P. Wang, J. D. Schmahmann, T. Benner, W. Y. Tseng et al., Diffusion spectrum magnetic resonance imaging (DSI) tractography of crossing fibers, Neuroimage, vol.41, pp.1267-1277, 2008.

E. Fieremans, J. H. Jensen, and J. A. Helpern, White matter characterization with diffusional kurtosis imaging, Neuroimage, vol.58, pp.177-188, 2011.
DOI : 10.1016/j.neuroimage.2011.06.006

URL : http://europepmc.org/articles/pmc3136876?pdf=render

E. S. Hui, M. M. Cheung, L. Qi, and E. X. Wu, Advanced MR diffusion characterization of neural tissue using directional diffusion kurtosis analysis, Conf Proc IEEE Eng Med Biol Soc, pp.3941-3944, 2008.
DOI : 10.1109/iembs.2008.4650072

J. H. Jensen and J. A. Helpern, Progress in diffusion-weighted imaging: concepts, techniques and applications to the central nervous system, NMR Biomed, vol.23, pp.659-660, 2010.

S. Lanzafame, M. Giannelli, F. Garaci, R. Floris, A. Duggento et al., Differences in Gaussian diffusion tensor imaging and non-Gaussian diffusion kurtosis imaging model-based estimates of diffusion tensor invariants in the human brain, Med Phys, vol.43, p.2464, 2016.

M. Giannelli and N. Toschi, On the use of trace-weighted images in body diffusional kurtosis imaging, Magn Reson Imaging, vol.34, pp.502-507, 2016.

D. C. Alexander, Multiple-fiber reconstruction algorithms for diffusion MRI, Ann N Y Acad Sci, vol.1064, pp.113-133, 2005.

Y. Assaf, T. Blumenfeld-katzir, Y. Yovel, and P. J. Basser, AxCaliber: a method for measuring axon diameter distribution from diffusion MRI, Magn Reson Med, vol.59, pp.1347-1354, 2008.

Y. Assaf and P. J. Basser, Composite hindered and restricted model of diffusion (CHARMED) MR imaging of the human brain, Neuroimage, vol.27, pp.48-58, 2005.

S. D. Santis, T. Granberg, R. Ouellette, C. A. Treaba, Q. Fan et al., 39th Annual International Conference of the IEEE Engineering in Medicine and Biology Society, pp.3024-3027, 2017.

D. C. Alexander, P. L. Hubbard, M. G. Hall, E. A. Moore, M. Ptito et al., Orientationally invariant indices of axon diameter and density from diffusion MRI, Neuroimage, vol.52, pp.1374-1389, 2010.

D. Santis, S. Gabrielli, A. Bozzali, M. Maraviglia, B. Macaluso et al., Anisotropic anomalous diffusion assessed in the human brain by scalar invariant indices, Magn Reson Med, vol.65, pp.1043-1052, 2011.

D. Santis, S. Gabrielli, A. Palombo, M. Maraviglia, B. Capuani et al., Non-Gaussian diffusion imaging: a brief practical review, Magn Reson Imaging, vol.29, pp.1410-1416, 2011.

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.

A. Iraji, E. Davoodi-bojd, H. Soltanian-zadeh, G. A. Hossein-zadeh, and Q. Jiang, Diffusion kurtosis imaging discriminates patients with white matter lesions from healthy subjects, Conf Proc IEEE Eng Med Biol Soc, pp.2796-2799, 2011.

M. F. Falangola, J. H. Jensen, J. S. Babb, C. Hu, F. X. Castellanos et al., Age-related non-Gaussian diffusion patterns in the prefrontal brain, J Magn Reson Imaging, vol.28, pp.1345-1350, 2008.

J. J. Wang, W. Y. Lin, C. S. Lu, Y. H. Weng, S. H. Ng et al., Parkinson disease: diagnostic utility of diffusion kurtosis imaging, Radiology, vol.261, pp.210-217, 2011.

P. M. Thompson, T. Ge, D. C. Glahn, N. Jahanshad, and T. E. Nichols, Genetics of the connectome. Neuroimage, vol.80, pp.475-488, 2013.

H. C. Baggio, B. Segura, C. Junque, M. A. De-reus, and R. Sala-llonch, Van den Heuvel MP. Rich club organization and cognitive performance in healthy older participants, J Cogn Neurosci, 2015.

T. T. Nakagawa, V. K. Jirsa, A. Spiegler, A. R. Mcintosh, and G. Deco, Bottom up modeling of the connectome: linking structure and function in the resting brain and their changes in aging, Neuroimage, vol.80, pp.318-329, 2013.

A. Fornito, A. Zalesky, and M. Breakspear, The connectomics of brain disorders, Nature Reviews Neuroscience, vol.16, pp.159-172, 2015.

F. U. Fischer, D. Wolf, A. Scheurich, A. I. Fellgiebel, and . Asdn, Altered whole-brain white matter networks in preclinical Alzheimer's disease, NeuroImage: Clinical, vol.8, pp.660-666, 2015.

M. Daianu, N. Jahanshad, T. M. Nir, A. W. Toga, C. R. Jack et al., Breakdown of brain connectivity between normal aging and Alzheimer's disease: a structural k-core network analysis, Brain connectivity, vol.3, pp.407-422, 2013.

N. K. Logothetis, J. Pauls, M. Augath, T. Trinath, and A. Oeltermann, Neurophysiological investigation of the basis of the fMRI signal, Nature, vol.412, pp.150-157, 2001.

M. M. Machulda, M. L. Senjem, S. D. Weigand, G. E. Smith, R. J. Ivnik et al., Functional magnetic resonance imaging changes in amnestic and nonamnestic mild cognitive impairment during encoding and recognition tasks, J Int Neuropsychol Soc, vol.15, p.372, 2009.

M. M. Machulda, H. A. Ward, B. Borowski, J. L. Gunter, R. H. Cha et al., Comparison of memory fMRI response among normal, MCI, and Alzheimer's patients, Neurology, vol.61, pp.500-506, 2003.

R. A. Sperling, fMRI studies of associative encoding in young and elderly controls and mild Alzheimer's disease, J Neurol Neurosurg Psychiatry, vol.74, pp.44-50, 2003.

J. P. Chhatwal and R. A. Sperling, Functional MRI of Mnemonic Networks Across the Spectrum of Normal Aging, Mild Cognitive Impairment, and Alzheimer's Disease, Journal of Alzheimers Disease, vol.31, pp.155-167, 2012.

B. C. Dickerson, S. L. Miller, D. N. Greve, A. M. Dale, M. S. Albert et al., Prefrontal-hippocampal-fusiform activity during encoding predicts intraindividual differences in free recall ability: An event-related functional-anatomic MRI study, Hippocampus, vol.17, pp.1060-1070, 2007.

K. V. Papp, R. E. Amariglio, E. C. Mormino, T. Hedden, M. Dekhytar et al., Free and cued memory in relation to biomarker-defined abnormalities in clinically normal older adults and those at risk for Alzheimer's disease, Neuropsychologia, vol.73, pp.169-175, 2015.

R. A. Sperling, B. C. Dickerson, M. Pihlajamaki, P. Vannini, P. S. Laviolette et al., Functional Alterations in Memory Networks in Early Alzheimer's Disease, Neuromolecular Med, vol.12, pp.27-43, 2010.

M. Pihlajamäki, K. O'keefe, L. Bertram, R. E. Tanzi, B. C. Dickerson et al., Evidence of Altered Posteromedial Cortical fMRI Activity in Subjects at Risk for Alzheimer Disease, Alzheimer Dis Assoc Disord, vol.24, pp.28-36, 2010.

S. Rombouts, R. Goekoop, C. J. Stam, F. Barkhof, and P. Scheltens, Delayed rather than decreased BOLD response as a marker for early Alzheimer's disease, Neuroimage, vol.26, pp.1078-1085, 2005.

K. A. Celone, V. D. Calhoun, B. C. Dickerson, A. Atri, E. F. Chua et al., Alterations in Memory Networks in Mild Cognitive Impairment and Alzheimer's Disease: An Independent Component Analysis, J Neurosci, vol.26, pp.10222-10231, 2006.

A. Bokde, M. Karmann, C. Born, S. J. Teipel, M. Omerovic et al., Altered Brain Activation During a Verbal Working Memory Task in Subjects with Amnestic Mild Cognitive Impairment, J Alzheimers Dis, vol.21, pp.103-118, 2010.

D. J. Veltman, S. Rombouts, and R. J. Dolan, Maintenance versus manipulation in verbal working memory revisited: an fMRI study, Neuroimage, vol.18, pp.247-256, 2003.

N. A. Kochan, M. Breakspear, M. Valenzuela, M. J. Slavin, H. Brodaty et al., Cortical Responses to a Graded Working Memory Challenge Predict Functional Decline in Mild Cognitive Impairment, Biol Psychiatry, vol.70, pp.123-130, 2011.

S. Teipel, I. Ehlers, A. Erbe, C. Holzmann, E. Lau et al., Structural Connectivity Changes Underlying Altered Working Memory Networks in Mild Cognitive Impairment: A Three-Way Image Fusion Analysis, J Neuroimaging, vol.25, pp.634-642, 2014.

D. J. Veltman, D. Ruiter, M. B. Rombouts, S. Lazeron, R. Barkhof et al., Hans Phaf R. Neurophysiological correlates of increased verbal working memory in highdissociative participants: a functional MRI study, Psychol Med, vol.35, pp.175-185, 2005.

N. A. Kochan, M. Breakspear, M. J. Slavin, M. Valenzuela, S. Mccraw et al., Functional Alterations in Brain Activation and Deactivation in Mild Cognitive Impairment in Response to a Graded Working Memory Challenge, Dement Geriatr Cogn Disord, vol.30, pp.553-568, 2010.

A. Bokde, W. Dong, C. Born, G. Leinsinger, T. Meindl et al., Task difficulty in a simultaneous face matching task modulates activity in face fusiform area, Cognitive Brain Research, vol.25, pp.701-710, 2005.

A. Bokde, S. J. Teipel, A. Drzezga, J. Thissen, P. Bartenstein et al., Association between Cognitive Performance and Cortical Glucose Metabolism in Patients with Mild Alzheimer's Disease, Dement Geriatr Cogn Disord, vol.20, pp.352-357, 2005.

D. M. Barch, T. S. Braver, L. E. Nystrom, S. D. Forman, D. C. Noll et al., Dissociating working memory from task difficulty in human prefrontal cortex, Neuropsychologia, vol.35, pp.1373-1380, 1997.

R. L. Gould, R. G. Brown, A. M. Owen, D. H. Ffytche, and R. J. Howard, FMRI BOLD response to increasing task difficulty during successful paired associates learning, Neuroimage, vol.20, pp.1006-1019, 2003.

C. L. Grady, B. Horwitz, P. Pietrini, M. J. Mentis, L. G. Ungerleider et al., Effect of task difficulty on cerebral blood flow during perceptual matching of faces, Hum Brain Mapp, vol.4, pp.227-239, 1996.

R. C. Gur, R. E. Gur, B. E. Skolnick, S. M. Resnick, F. L. Silver et al., Effects of Task Difficulty on Regional Cerebral Blood Flow: Relationships with Anxiety and Performance, Psychophysiology, vol.25, pp.392-399, 1988.

A. Bokde, P. Lopez-bayo, C. Born, M. Ewers, T. Meindl et al., Alzheimer Disease: Functional Abnormalities in the Dorsal Visual Pathway, Radiology, vol.254, pp.219-226, 2010.

A. Bokde, P. Lopez-bayo, C. Born, W. Dong, T. Meindl et al., Functional abnormalities of the visual processing system in subjects with mild cognitive impairment: An fMRI study, Psychiatry Research: Neuroimaging, vol.163, pp.248-259, 2008.

J. Hao, K. Li, K. Li, D. Zhang, W. Wang et al., Visual attention deficits in Alzheimer's disease: an fMRI study, Neurosci Lett, vol.385, pp.18-23, 2005.

M. L. Keightley, G. Winocur, S. J. Graham, H. S. Mayberg, S. J. Hevenor et al., An fMRI study investigating cognitive modulation of brain regions associated with emotional processing of visual stimuli, Neuropsychologia, vol.41, pp.585-596, 2003.

E. Matsumoto, Y. Ohigashi, M. Fujimori, and E. Mori, The Processing of Global and Local Visual Information in Alzheimer's Disease, Behav Neurol, vol.12, pp.119-125, 2000.

M. J. Mentis, G. E. Alexander, J. Krasuski, P. Pietrini, M. L. Furey et al., Increasing required neural response to expose abnormal brain function in mild versus moderate or severe Alzheimer's disease: PET study using parametric visual stimulation, Am J Psychiatry, vol.155, pp.785-794, 1998.

M. J. Mentis, B. Horwitz, C. L. Grady, G. E. Alexander, J. W. Vanmeter et al., Visual cortical dysfunction in Alzheimer's disease evaluated with a temporally graded "stress test" during PET, Am J Psychiatry, vol.153, pp.32-40, 1996.

P. Pietrini, G. E. Alexander, M. L. Furey, A. Dani, M. J. Mentis et al., Cerebral metabolic response to passive audiovisual stimulation in patients with Alzheimer's disease and healthy volunteers assessed by PET, J Nucl Med, vol.41, pp.575-583, 2000.

A. R. Mcintosh, C. L. Grady, L. G. Ungerleider, J. V. Haxby, S. I. Rapoport et al., Network analysis of cortical visual pathways mapped with PET, J Neurosci, vol.14, pp.655-666, 1994.

P. Pietrini, M. L. Furey, N. Graff-radford, U. Freo, G. E. Alexander et al., Preferential metabolic involvement of visual cortical areas in a subtype of

, Alzheimer's disease: clinical implications, Am J Psychiatry, vol.153, pp.1261-1268, 1996.

D. Prvulovic, D. Hubl, A. T. Sack, L. Melillo, K. Maurer et al., Functional Imaging of Visuospatial Processing in Alzheimer's Disease, Neuroimage, vol.17, pp.1403-1414, 2002.

A. Bokde, E. Cavedo, P. Lopez-bayo, S. Lista, T. Meindl et al., Effects of rivastigmine on visual attention in subjects with amnestic mild cognitive impairment: A serial functional MRI activation pilot-study, Psychiatry Research: Neuroimaging, vol.249, pp.84-90, 2016.

A. Hämäläinen, M. Pihlajamäki, H. Tanila, T. Hänninen, E. Niskanen et al., Increased fMRI responses during encoding in mild cognitive impairment, Neurobiol Aging, vol.28, pp.1889-1903, 2007.

P. Bentley, J. Driver, and R. J. Dolan, Cholinesterase inhibition modulates visual and attentional brain responses in Alzheimer's disease and health, Brain, vol.131, pp.409-424, 2008.

P. Bentley, J. Driver, and R. J. Dolan, Modulation of fusiform cortex activity by cholinesterase inhibition predicts effects on subsequent memory, Brain, vol.132, pp.2356-2371, 2009.

P. Bentley, M. Husain, and R. J. Dolan, Effects of Cholinergic Enhancement on Visual Stimulation, Spatial Attention, and Spatial Working Memory, Neuron, vol.41, pp.969-982, 2004.

A. Bokde, M. Karmann, S. J. Teipel, C. Born, M. Lieb et al., Decreased Activation Along the Dorsal Visual Pathway After a 3-Month Treatment With Galantamine in Mild Alzheimer Disease, J Clin Psychopharmacol, vol.29, pp.147-156, 2009.

J. H. Balsters, O. Connell, R. G. Martin, M. P. Galli, A. Cassidy et al., Donepezil Impairs Memory in Healthy Older Subjects: Behavioural, EEG and Simultaneous EEG/fMRI Biomarkers, vol.6, 2011.

P. Bentley, P. Vuilleumier, C. M. Thiel, J. Driver, and R. J. Dolan, Cholinergic enhancement modulates neural correlates of selective attention and emotional processing, Neuroimage, vol.20, pp.58-70, 2003.

G. Handjaras, E. Ricciardi, J. Szczepanik, P. Pietrini, and M. L. Furey, Cholinergic enhancement differentially modulates neural response to encoding during face identity and face location working memory tasks, Exp Biol Med, vol.238, pp.999-1008, 2013.

E. Ricciardi, G. Handjaras, G. Bernardi, P. Pietrini, and M. L. Furey, Cholinergic enhancement reduces functional connectivity and BOLD variability in visual extrastriate cortex during selective attention, Neuropharmacology, vol.64, pp.305-313, 2013.

C. E. Pizoli, M. N. Shah, A. Z. Snyder, J. S. Shimony, D. D. Limbrick et al., Resting-state activity in development and maintenance of normal brain function, Proceedings of the National Academy of Sciences, vol.108, pp.11638-11643, 2011.

S. Chen, T. J. Ross, W. Zhan, C. S. Myers, K. Chuang et al., Group independent component analysis reveals consistent resting-state networks across multiple sessions, Brain Res, vol.1239, pp.141-151, 2008.

M. D. Greicius, B. Krasnow, A. L. Reiss, and V. Menon, Functional connectivity in the resting brain: A network analysis of the default mode hypothesis, Proceedings of the National Academy of Sciences, vol.100, pp.253-258, 2002.

M. D. Greicius, K. Supekar, V. Menon, and R. F. Dougherty, Resting-State Functional Connectivity Reflects Structural Connectivity in the Default Mode Network, Cereb Cortex, vol.19, pp.72-78, 2008.

L. M. Shah, J. A. Cramer, M. A. Ferguson, R. M. Birn, and J. S. Anderson, Reliability and reproducibility of individual differences in functional connectivity acquired during task and resting state, Brain and behavior, p.6, 2016.

R. M. Hutchison, T. Womelsdorf, E. A. Allen, P. A. Bandettini, V. D. Calhoun et al., Dynamic functional connectivity: promise, issues, and interpretations, Neuroimage, vol.80, pp.360-378, 2013.

D. Power-jonathan, C. Alexander, L. , N. Steven, M. et al., Functional Network Organization of the Human Brain, Neuron, vol.72, pp.665-678, 2011.

W. Wen, W. Zhu, Y. He, N. A. Kochan, S. Reppermund et al., Discrete Neuroanatomical Networks Are Associated with Specific Cognitive Abilities in Old Age, J Neurosci, vol.31, pp.1204-1212, 2011.

M. W. Cole, D. S. Bassett, J. D. Power, T. S. Braver, and S. E. Petersen, Intrinsic and task-evoked network architectures of the human brain, Neuron, vol.83, pp.238-251, 2014.

J. Connolly, J. P. Mcnulty, L. Boran, R. Roche, D. Delany et al., Identification of Resting State Networks Involved in Executive Function, Brain Connectivity, vol.6, pp.365-374, 2016.

R. Sala-llonch, C. Peña-gómez, E. M. Arenaza-urquijo, D. Vidal-piñeiro, N. Bargalló et al., Bartrés-Faz D. Brain connectivity during resting state and subsequent working memory task predicts behavioural performance, Cortex, vol.48, pp.1187-1196, 2012.

G. Wang, T. G. Belgard, D. Mao, L. Chen, S. Berto et al., Correspondence between Resting-State Activity and Brain Gene Expression, Neuron, vol.88, pp.659-666, 2015.

J. Richiardi, A. Altmann, A. C. Milazzo, C. Chang, M. M. Chakravarty et al., Consortium I. Correlated gene expression supports synchronous activity in brain networks, Science, vol.348, pp.1241-1244, 2015.

R. C. Craddock, P. E. Holtzheimer, X. P. Hu, and H. S. Mayberg, Disease state prediction from resting state functional connectivity, Magn Reson Med, vol.62, pp.1619-1628, 2009.

M. D. Greicius, G. Srivastava, A. L. Reiss, and V. Menon, Default-mode network activity distinguishes Alzheimer's disease from healthy aging: Evidence from functional MRI, Proceedings of the National Academy of Sciences, vol.101, pp.4637-4642, 2004.

W. W. Seeley, R. K. Crawford, J. Zhou, B. L. Miller, and M. D. Greicius, Neurodegenerative Diseases Target Large-Scale Human Brain Networks, Neuron, vol.62, pp.42-52, 2009.

W. W. Seeley, V. Menon, A. F. Schatzberg, J. Keller, G. H. Glover et al., Dissociable Intrinsic Connectivity Networks for Salience Processing and Executive Control, J Neurosci, vol.27, pp.2349-2356, 2007.

J. Zhou, M. D. Greicius, E. D. Gennatas, M. E. Growdon, J. Y. Jang et al., Divergent network connectivity changes in behavioural variant frontotemporal dementia and Alzheimer's disease, Brain, vol.133, pp.1352-1367, 2010.

E. G. Dopper, S. A. Rombouts, L. C. Jiskoot, T. Den-heijer, J. R. De-graaf et al., Structural and functional brain connectivity in presymptomatic familial frontotemporal dementia

J. B. Thomas, M. R. Brier, R. J. Bateman, A. Z. Snyder, T. L. Benzinger et al., Functional Connectivity in Autosomal Dominant and Late-Onset Alzheimer Disease, JAMA Neurology, vol.71, p.1111, 2014.

A. M. Ward, E. C. Mormino, W. Huijbers, A. P. Schultz, T. Hedden et al., Relationships between default-mode network connectivity, medial temporal lobe structure, and age-related memory deficits, Neurobiol Aging, vol.36, pp.265-272, 2015.

M. W. Cole, T. Ito, D. S. Bassett, and D. H. Schultz, Activity flow over resting-state networks shapes cognitive task activations, Nat Neurosci, vol.19, pp.1718-1726, 2016.

M. Mennes, C. Kelly, X. N. Zuo, D. Martino, A. Biswal et al., Interindividual differences in resting-state functional connectivity predict task-induced BOLD activity, Neuroimage, vol.50, pp.1690-1701, 2010.

M. Mennes, X. Zuo, C. Kelly, D. Martino, A. Zang et al., Linking inter-individual differences in neural activation and behavior to intrinsic brain dynamics, Neuroimage, vol.54, pp.2950-2959, 2011.

M. Lorenzi, A. Beltramello, N. B. Mercuri, E. Canu, G. Zoccatelli et al., Effect of Memantine on Resting State Default Mode Network Activity in Alzheimer's Disease, Drugs Aging, vol.28, pp.205-217, 2011.

T. O. Laumann, E. M. Gordon, B. Adeyemo, A. Z. Snyder, S. J. Joo et al., Functional system and areal organization of a highly sampled individual human brain, Neuron, vol.87, pp.657-670, 2015.

J. Dubois and R. Adolphs, Building a Science of Individual Differences from fMRI, Trends in Cognitive Sciences, vol.20, pp.425-443, 2016.

C. Babiloni, R. Lizio, N. Marzano, P. Capotosto, A. Soricelli et al., Brain neural synchronization and functional coupling in Alzheimer's disease as revealed by resting state EEG rhythms, Int J Psychophysiol, vol.103, pp.88-102, 2016.

Y. Stern, An approach to studying the neural correlates of reserve, Brain Imaging Behav, vol.11, pp.410-416, 2017.

C. Babiloni, D. Percio, C. Boccardi, M. Lizio, R. Lopez et al., Occipital sources of resting-state alpha rhythms are related to local gray matter density in subjects with amnesic mild cognitive impairment and Alzheimer's disease, Neurobiol Aging, vol.36, pp.556-570, 2015.

T. Dierks, R. Ihl, L. Frölich, and K. Maurer, Dementia of the alzheimer type: Effects on the spontaneous EEG described by dipole sources, Psychiatry Research: Neuroimaging, vol.50, pp.151-162, 1993.

T. Dierks, V. Jelic, R. D. Pascual-marqui, L. Wahlund, P. Julin et al., Spatial pattern of cerebral glucose metabolism (PET) correlates with localization of intracerebral EEG-generators in Alzheimer's disease, Clin Neurophysiol, vol.111, pp.1817-1824, 2000.

C. Huang, L. O. Wahlund, T. Dierks, P. Julin, B. Winblad et al., Discrimination of Alzheimer's disease and mild cognitive impairment by equivalent EEG sources: a cross-sectional and longitudinal study, Clin Neurophysiol, vol.111, pp.1961-1967, 2000.

V. Jelic, S. E. Johansson, O. Almkvist, M. Shigeta, P. Julin et al., Quantitative electroencephalography in mild cognitive impairment: longitudinal changes and possible prediction of Alzheimer's disease, Neurobiol Aging, vol.21, pp.533-540, 2000.

N. V. Ponomareva, N. D. Selesneva, and G. A. Jarikov, EEG Alterations in Subjects at High Familial Risk for Alzheimer's Disease, Neuropsychobiology, vol.48, pp.152-159, 2003.

J. Jeong, EEG dynamics in patients with Alzheimer's disease, Clin Neurophysiol, vol.115, pp.1490-1505, 2004.

C. Babiloni, G. Binetti, E. Cassetta, G. D. Forno, C. D. Percio et al., Sources of cortical rhythms change as a function of cognitive impairment in pathological aging: a multicenter study, Clin Neurophysiol, vol.117, pp.252-268, 2006.

R. R. Reeves, F. A. Struve, and G. Patrick, The Effects of Donepezil on Quantitative EEG in Patients with Alzheimer's Disease, Clin Electroencephalogr, vol.33, pp.93-96, 2002.

S. Brassen and G. Adler, Short-term effects of acetylcholinesterase inhibitor treatment on EEG and memory performance in Alzheimer patients: an open, controlled trial, Pharmacopsychiatry, vol.36, pp.304-308, 2003.

M. Onofrj, A. Thomas, D. Iacono, A. L. Luciano, D. Iorio et al., The Effects of a Cholinesterase Inhibitor Are Prominent in Patients With Fluctuating Cognition: A Part 3 Study of the Main Mechanism of Cholinesterase Inhibitors in Dementia, Clin Neuropharmacol, vol.26, pp.239-251, 2003.

E. A. Kogan, A. D. Korczyn, R. G. Virchovsky, S. S. Klimovizky, T. A. Treves et al., EEG changes during long-term treatment with donepezil in Alzheimer's disease patients, J Neural Transm, vol.108, pp.1167-1173, 2001.

G. Rodriguez, P. Vitali, C. De-leo, D. Carli, F. Girtler et al., Quantitative EEG Changes in Alzheimer Patients during Long-Term Donepezil Therapy, Neuropsychobiology, vol.46, pp.49-56, 2002.

D. C. Valladares-neto, M. S. Buchsbaum, W. J. Evans, D. Nguyen, P. Nguyen et al., EEG delta, positron emission tomography, and memory deficit in Alzheimer's disease, Neuropsychobiology, vol.31, pp.173-181, 1995.

C. Babiloni, D. Percio, C. Lizio, R. Noce, G. Cordone et al., Abnormalities of cortical neural synchronization mechanisms in patients with dementia due to Alzheimer's and Lewy body diseases: an EEG study, Neurobiol Aging, vol.55, pp.143-158, 2017.

G. Adler, S. Brassen, and A. Jajcevic, EEG coherence in Alzheimer's dementia, J Neural Transm, vol.110, pp.1051-1058, 2003.

R. Anghinah, P. Kanda, M. S. Jorge, E. Lima, L. Pascuzzi et al., Estudo da coerência do eletrencefalograma para a banda de frequência alfa em indivíduos adultos normais e com provável demência do tipo Alzheimer, Arq Neuropsiquiatr, vol.58, pp.272-275, 2000.

C. Besthorn, H. Förstl, C. Geiger-kabisch, H. Sattel, T. Gasser et al., EEG coherence in Alzheimer disease, Electroencephalogr Clin Neurophysiol, vol.90, pp.242-245, 1994.

J. J. Dunkin, A. F. Leuchter, T. F. Newton, and I. A. Cook, Reduced EEG coherence in dementia: State or trait marker?, Biol Psychiatry, vol.35, pp.870-879, 1994.

L. C. Fonseca, G. Tedrus, L. R. Prandi, A. M. Almeida, and D. S. Furlanetto, Alzheimer's disease: relationship between cognitive aspects and power and coherence EEG measures, Arq Neuropsiquiatr, vol.69, pp.875-881, 2011.

L. C. Fonseca, G. Tedrus, P. N. Carvas, and E. Machado, Comparison of quantitative EEG between patients with Alzheimer's disease and those with Parkinson's disease dementia, Clin Neurophysiol, vol.124, pp.1970-1974, 2013.

V. Jelic, P. Julin, M. Shigeta, A. Nordberg, L. Lannfelt et al., Apolipoprotein E epsilon 4 allele decreases functional connectivity in Alzheimer's disease as measured by EEG coherence, J Neurol Neurosurg Psychiatry, vol.63, pp.59-65, 1997.

V. Knott, C. Engeland, E. Mohr, C. Mahoney, and V. Ilivitsky, Acute Nicotine Administration in Alzheimer's Disease: An Exploratory EEG Study, Neuropsychobiology, vol.41, pp.210-220, 2000.

A. F. Leuchter, Electroencephalographic Spectra and Coherence in the Diagnosis of Alzheimer'sType and Multi-infarct Dementia, Arch Gen Psychiatry, vol.44, p.993, 1987.

A. F. Leuchter, T. F. Newton, I. A. Cook, D. O. Walter, S. Rosenberg-thompson et al., Brain, vol.115, pp.1543-1561, 1992.

T. Locatelli, M. Cursi, D. Liberati, M. Franceschi, and G. Comi, EEG coherence in Alzheimer's disease, Electroencephalogr Clin Neurophysiol, vol.106, pp.229-237, 1998.

O. Pogarell, EEG coherence reflects regional corpus callosum area in Alzheimer's disease, J Neurol Neurosurg Psychiatry, vol.76, pp.109-111, 2005.
DOI : 10.1136/jnnp.2004.036566

URL : https://jnnp.bmj.com/content/76/1/109.full.pdf

E. P. Sloan, G. W. Fenton, N. Kennedy, and J. M. Maclennan, Neurophysiology and SPECT cerebral blood flow patterns in dementia, Electroencephalogr Clin Neurophysiol, vol.91, pp.163-170, 1994.
DOI : 10.1016/0013-4694(94)90066-3

A. F. Leuchter, J. J. Dunkin, R. B. Lufkin, Y. Anzai, I. A. Cook et al., Effect of white matter disease on functional connections in the aging brain, J Neurol Neurosurg Psychiatry, vol.57, pp.1347-1354, 1994.

C. Babiloni, G. B. Frisoni, F. Vecchio, M. Pievani, C. Geroldi et al., Global Functional Coupling of Resting EEG Rhythms is Related to White-Matter Lesions Along the Cholinergic Tracts in Subjects with Amnesic Mild Cognitive Impairment, J Alzheimers Dis, vol.19, pp.859-871, 2010.

Z. Sankari, H. Adeli, and A. Adeli, Intrahemispheric, interhemispheric, and distal EEG coherence in Alzheimer's disease, Clin Neurophysiol, vol.122, pp.897-906, 2011.
DOI : 10.1016/j.clinph.2010.09.008

C. Babiloni, F. Babiloni, F. Carducci, F. Cincotti, F. Vecchio et al., Functional Frontoparietal Connectivity During Short-Term Memory as Revealed by HighResolution EEG Coherence Analysis, Behav Neurosci, vol.118, pp.687-697, 2004.
DOI : 10.1037/0735-7044.118.4.687

C. J. Stam, G. Nolte, and A. Daffertshofer, Phase lag index: Assessment of functional connectivity from multi channel EEG and MEG with diminished bias from common sources, Hum Brain Mapp, vol.28, pp.1178-1193, 2007.

W. De-haan, W. M. Van-der-flier, H. Wang, P. Van-mieghem, P. Scheltens et al., Disruption of Functional Brain Networks in Alzheimer's Disease: What Can We Learn from Graph Spectral Analysis of Resting-State Magnetoencephalography?, Brain Connectivity, vol.2, pp.45-55, 2012.

F. Vecchio, F. Miraglia, C. Marra, D. Quaranta, M. G. Vita et al., Human Brain Networks in Cognitive Decline: A Graph Theoretical Analysis of Cortical Connectivity from EEG Data, Journal of Alzheimers Disease, vol.41, pp.113-127, 2014.

F. C. Morabito, M. Campolo, D. Labate, G. Morabito, L. Bonanno et al., A Longitudinal EEG Study of Alzheimer's Disease Progression Based on A Complex Network Approach, Int J Neural Syst, vol.25, p.1550005, 2015.

V. Drago, C. Babiloni, D. Bartres-faz, A. Caroli, B. Bosch et al., Disease Tracking Markers for Alzheimer's Disease at the Prodromal (MCI) Stage, Journal of Alzheimers Disease, vol.26, pp.159-199, 2011.
DOI : 10.3233/jad-2011-0043

L. A. Coben, W. Danziger, and M. Storandt, A longitudinal EEG study of mild senile dementia of Alzheimer type: changes at 1 year and at 2.5 years, Electroencephalogr Clin Neurophysiol, vol.61, pp.101-112, 1985.

H. Soininen, J. Partanen, V. Laulumaa, E. L. Helkala, M. Laakso et al., Longitudinal EEG spectral analysis in early stage of Alzheimer's disease, Electroencephalogr Clin Neurophysiol, vol.72, pp.290-297, 1989.
DOI : 10.1016/0013-4694(89)90064-3

H. Soininen, J. Partanen, V. Laulumaa, A. Pääkkönen, E. L. Helkala et al., Serial EEG in Alzheimer's disease: 3 year follow-up and clinical outcome, Electroencephalogr Clin Neurophysiol, vol.79, pp.342-348, 1991.
DOI : 10.1016/0013-4694(91)90198-d

C. Babiloni, D. Bartrés-faz, M. Didic, G. Forloni, J. Jovicich et al., Cross-sectional clinical, neuropsychological, neuroimaging, neurophysiological and biochemical characterization of people with mild cognitive impairment in the WP5 PharmaCog/E-ADNI study, Alzheimer's & Dementia, vol.9, p.48, 2013.

C. Babiloni, D. Percio, C. Lizio, R. Marzano, N. Infarinato et al., Cortical sources of resting state electroencephalographic alpha rhythms deteriorate across time in subjects with amnesic mild cognitive impairment, Neurobiol Aging, vol.35, pp.130-142, 2014.

G. Adler and S. Brassen, Short-Term Rivastigmine Treatment Reduces EEG Slow-Wave Power in Alzheimer Patients, Neuropsychobiology, vol.43, pp.273-276, 2001.
DOI : 10.1159/000054902

S. Balkan, N. Yaras, E. Mihci, B. Dora, A. Agar et al., Effect of donepezil on EEG spectral analysis in Alzheimer's disease, Acta Neurol Belg, vol.103, pp.164-169, 2003.

L. Gianotti, G. Künig, P. L. Faber, D. Lehmann, R. D. Pascual-marqui et al., Rivastigmine effects on EEG spectra and three-dimensional LORETA functional imaging in Alzheimer's disease, Psychopharmacology (Berl), vol.198, pp.323-332, 2008.
DOI : 10.1007/s00213-008-1111-1

URL : http://doc.rero.ch/record/309638/files/213_2008_Article_1111.pdf

G. Adler, S. Brassen, K. Chwalek, B. Dieter, and M. Teufel, Prediction of treatment response to rivastigmine in Alzheimer's dementia, Journal of Neurology Neurosurgery and Psychiatry, vol.75, pp.292-294, 2004.

A. Agnoli, N. Martucci, V. Manna, L. Conti, and M. Fioravanti, Effect of Cholinergic and Anticholinergic Drugs on Short-Term Memory in Alzheimer's Dementia, Clin Neuropharmacol, vol.6, pp.311-324, 1983.

K. Alhainen, J. Partanen, K. Reinikainen, V. Laulumaa, H. Soininen et al., Discrimination of tetrahydroaminoacridine responders by a single dose pharmaco-EEG in patients with Alzheimer's disease, Neurosci Lett, vol.127, pp.113-116, 1991.

K. Alhainen and P. J. Riekkinen, Discrimination of Alzheimer Patients Responding to Cholinesterase Inhibitor Therapy, Acta Neurol Scand, vol.88, pp.16-21, 1993.

G. Rodriguez, P. Vitali, M. Canfora, P. Calvini, N. Girtler et al., Quantitative EEG and perfusional single photon emission computed tomography correlation during long-term donepezil therapy in Alzheimer's disease, Clin Neurophysiol, vol.115, pp.39-49, 2004.

T. Kai, Y. Asai, K. Sakuma, T. Koeda, and K. Nakashima, Quantitative electroencephalogram analysis in dementia with Lewy bodies and Alzheimer's disease, J Neurol Sci, vol.237, pp.89-95, 2005.

S. Passero, R. Rocchi, G. Vatti, L. Burgalassi, and N. Battistini, Quantitative EEG Mapping, Regional Cerebral Blood Flow, and Neuropsychological Function in Alzheimer's Disease, Dement Geriatr Cogn Disord, vol.6, pp.148-156, 1995.

D. C. Valladares-neto, M. S. Buchsbaum, J. Evans, D. Nguyen, P. Nguyen et al., Positron Emission Tomography, and Memory Deficit in Alzheimer's Disease, Neuropsychobiology, vol.31, pp.173-181, 2008.

E. Niedermeyer, Alpha rhythms as physiological and abnormal phenomena, Int J Psychophysiol, vol.26, pp.31-49, 1997.

G. Rodriguez, F. Nobili, F. Copello, P. Vitali, M. V. Gianelli et al., 99mTc-HMPAO regional cerebral blood flow and quantitative electroencephalography in Alzheimer's disease: a correlative study, J Nucl Med, vol.40, pp.522-529, 1999.

A. Fernandez, J. Arrazola, F. Maestu, C. Amo, P. Gil-gregorio et al., Correlations of hippocampal atrophy and focal low-frequency magnetic activity in Alzheimer disease: volumetric MR imaging-magnetoencephalographic study, AJNR Am J Neuroradiol, vol.24, pp.481-487, 2003.

N. R. Graff-radford, L. M. Besser, J. E. Crook, W. A. Kukull, and D. W. Dickson, Neuropathologic differences by race from the National Alzheimer's Coordinating Center, vol.12, pp.669-677, 2016.

L. Sarro, M. L. Senjem, E. S. Lundt, S. A. Przybelski, T. G. Lesnick et al., Amyloid-? deposition and regional grey matter atrophy rates in dementia with Lewy bodies, Brain, vol.139, pp.2740-2750, 2016.

S. W. Hughes and V. Crunelli, Thalamic Mechanisms of EEG Alpha Rhythms and Their Pathological Implications, The Neuroscientist, vol.11, pp.357-372, 2005.

M. L. Lorincz, V. Crunelli, and S. W. Hughes, Cellular Dynamics of Cholinergically Induced (8 13 Hz) Rhythms in Sensory Thalamic Nuclei In Vitro, J Neurosci, vol.28, pp.660-671, 2008.

M. L. L?rincz, K. A. Kékesi, G. Juhász, V. Crunelli, and S. W. Hughes, Temporal Framing of Thalamic Relay-Mode Firing by Phasic Inhibition during the Alpha Rhythm, Neuron, vol.63, pp.683-696, 2009.

M. Andersson, O. Hansson, L. Minthon, I. Rosén, and E. Londos, Electroencephalogram Variability in Dementia with Lewy Bodies, Dement Geriatr Cogn Disord, vol.26, pp.284-290, 2008.

K. Engedal, J. Snaedal, P. Hoegh, V. Jelic, B. Andersen et al., Quantitative EEG Applying the Statistical Recognition Pattern Method: A Useful Tool in Dementia Diagnostic Workup, Dement Geriatr Cogn Disord, vol.40, pp.1-12, 2015.

H. Garn, C. Coronel, M. Waser, G. Caravias, and G. Ransmayr, Differential diagnosis between patients with probable Alzheimer's disease, Parkinson's disease dementia, or dementia with Lewy bodies and frontotemporal dementia, behavioral variant, using quantitative electroencephalographic features, J Neural Transm, vol.124, pp.569-581, 2017.

M. Dauwan, J. J. Van-der-zande, E. Van-dellen, I. Sommer, P. Scheltens et al., Random forest to differentiate dementia with Lewy bodies from Alzheimer's disease

, Alzheimer's & Dementia: Diagnosis, Assessment & Disease Monitoring, vol.4, pp.99-106, 2016.

C. Babiloni, D. Percio, C. Lizio, R. Noce, G. Cordone et al., Abnormalities of Cortical Neural Synchronization Mechanisms in Subjects with Mild Cognitive Impairment due to Alzheimer's and Parkinson's Diseases: An EEG Study, J Alzheimers Dis, vol.59, pp.339-358, 2017.

P. M. Rossini, D. Percio, C. Pasqualetti, P. Cassetta, E. Binetti et al., Conversion from mild cognitive impairment to Alzheimer's disease is predicted by sources and coherence of brain electroencephalography rhythms, Neuroscience, vol.143, pp.793-803, 2006.

K. Van-der-hiele, E. Bollen, A. A. Vein, R. Reijntjes, R. Westendorp et al., EEG Markers of Future Cognitive Performance in the Elderly, J Clin Neurophysiol, vol.25, pp.83-89, 2008.

C. Luckhaus, B. Grass-kapanke, I. Blaeser, R. Ihl, T. Supprian et al., Quantitative EEG in progressingvsstable mild cognitive impairment (MCI): results of a 1-year follow-up study, Int J Geriatr Psychiatry, vol.23, pp.1148-1155, 2008.

P. Walla, E. Püregger, J. Lehrner, D. Mayer, L. Deecke et al., Depth of word processing in Alzheimer patients and normal controls: a magnetoencephalographic (MEG) study, J Neural Transm, vol.112, pp.713-730, 2004.

M. I. Garrido, J. M. Kilner, K. E. Stephan, and K. J. Friston, The mismatch negativity: A review of underlying mechanisms, Clin Neurophysiol, vol.120, pp.453-463, 2009.

R. Näätänen, P. Paavilainen, T. Rinne, and K. Alho, The mismatch negativity (MMN) in basic research of central auditory processing: A review, Clin Neurophysiol, vol.118, pp.2544-2590, 2007.

C. Cheng, P. Wang, W. Hsu, and Y. Lin, Inadequate inhibition of redundant auditory inputs in Alzheimer's disease: An MEG study, Biol Psychol, vol.89, pp.365-373, 2012.

E. Pekkonen, Mismatch Negativity in Aging and in Alzheimer's and Parkinson's Diseases, Audiology and Neurotology, vol.5, pp.216-224, 2000.

S. P. Deeny, D. Poeppel, J. B. Zimmerman, S. M. Roth, J. Brandauer et al., Exercise, APOE, and working memory: MEG and behavioral evidence for benefit of exercise in epsilon4 carriers, Biol Psychol, vol.78, pp.179-187, 2008.

F. Maestú, P. Campo, P. Gil-gregorio, S. Fernández, A. Fernández et al., Medial temporal lobe neuromagnetic hypoactivation and risk for developing cognitive decline in elderly population: A 2-year follow-up study, Neurobiol Aging, vol.27, pp.32-37, 2006.

C. J. Stam, Use of magnetoencephalography (MEG) to study functional brain networks in neurodegenerative disorders, J Neurol Sci, vol.289, pp.128-134, 2010.

M. E. Raichle, A. M. Macleod, A. Z. Snyder, W. J. Powers, D. A. Gusnard et al., A default mode of brain function, Proceedings of the National Academy of Sciences, vol.98, pp.676-682, 2001.

M. J. Brookes, J. R. Hale, J. M. Zumer, C. M. Stevenson, S. T. Francis et al., Measuring functional connectivity using MEG: Methodology and comparison with fcMRI, Neuroimage, vol.56, pp.1082-1104, 2011.
DOI : 10.1016/j.neuroimage.2011.02.054

URL : https://doi.org/10.1016/j.neuroimage.2011.02.054

M. J. Brookes, P. K. Tewarie, B. Hunt, S. E. Robson, L. E. Gascoyne et al., A multi-layer network approach to MEG connectivity analysis, Neuroimage, vol.132, pp.425-438, 2016.

L. Marzetti, D. Penna, S. Snyder, A. Z. Pizzella, V. Nolte et al., Frequency specific interactions of MEG resting state activity within and across brain networks as revealed by the multivariate interaction measure, Neuroimage, vol.79, pp.172-183, 2013.

F. De-pasquale, D. Penna, S. Snyder, A. Z. Lewis, C. Mantini et al., Temporal dynamics of spontaneous MEG activity in brain networks, Proceedings of the National Academy of Sciences, vol.107, pp.6040-6045, 2010.

F. De-pasquale, D. Penna, S. Sporns, O. Romani, G. L. Corbetta et al., A Dynamic Core Network and Global Efficiency in the Resting Human Brain, Cereb Cortex, vol.26, pp.4015-4033, 2015.

P. Tewarie, A. Hillebrand, B. W. Van-dijk, C. J. Stam, O. Neill et al., Integrating cross-frequency and within band functional networks in resting-state MEG: A multi-layer network approach, Neuroimage, vol.142, pp.324-336, 2016.

S. Sockeel, D. Schwartz, M. Pélégrini-issac, and H. Benali, Large-Scale Functional Networks Identified from Resting-State EEG Using Spatial ICA, PLoS One, vol.11, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01274101

A. Besga, L. Ortiz, A. Fernández, F. Maestu, J. Arrazola et al., Structural and Functional Patterns in Healthy Aging, Mild Cognitive Impairment, and Alzheimer Disease, Alzheimer Dis Assoc Disord, vol.24, pp.1-10, 2010.

A. Fernández, R. Hornero, A. Mayo, J. Poza, P. Gil-gregorio et al., MEG spectral profile in Alzheimer's disease and mild cognitive impairment, Clin Neurophysiol, vol.117, pp.306-314, 2006.

A. Fernández, T. An, P. Zuluaga, P. Gil-gregorio, F. Maestu et al., Alzheimer's & Dementia, vol.2, p.673, 2006.

A. Fernandez, A. Turrero, P. Zuluaga, P. Gil-gregorio, F. Del-pozo et al., Mapping Along the Healthy Aging-Alzheimer's Disease Continuum: Diagnostic Implications, Journal of Alzheimers Disease, vol.35, pp.495-507, 2013.

W. De-haan, C. J. Stam, B. F. Jones, I. Zuiderwijk, B. W. Van-dijk et al., P1-388: Resting-state oscillatory brain dynamics in Alzheimer's disease, Alzheimer's & Dementia, vol.4, pp.332-333, 2008.

D. Osipova, K. Rantanen, J. Ahveninen, R. Ylikoski, O. Häppölä et al., Source estimation of spontaneous MEG oscillations in mild cognitive impairment, Neurosci Lett, vol.405, pp.57-61, 2006.

J. Guillon, Y. Attal, O. Colliot, L. Corte, V. Dubois et al., Loss of brain inter-frequency hubs in Alzheimer's disease. Sci Rep, 2017.

M. E. López, P. Cuesta, P. Garcés, P. N. Castellanos, S. Aurtenetxe et al., MEG spectral analysis in subtypes of mild cognitive impairment, AGE, p.36, 2014.

E. Zamrini, F. Maestu, E. Pekkonen, M. Funke, J. Makela et al., Magnetoencephalography as a Putative Biomarker for Alzheimer's Disease, Int J Alzheimers Dis, vol.2011, pp.1-10, 2011.

S. D. Hall, G. R. Barnes, P. L. Furlong, S. Seri, and A. Hillebrand, Neuronal network pharmacodynamics of GABAergic modulation in the human cortex determined using pharmacomagnetoencephalography, Hum Brain Mapp, vol.31, pp.581-594, 2010.

S. D. Muthukumaraswamy, The use of magnetoencephalography in the study of psychopharmacology (pharmaco-MEG), Journal of Psychopharmacology, vol.28, pp.815-829, 2014.

P. Cuesta, A. Barabash, S. Aurtenetxe, P. Garces, M. E. Lopez et al., Source Analysis of Spontaneous Magnetoencephalograpic Activity in Healthy Aging and Mild Cognitive Impairment: Influence of Apolipoprotein E Polymorphism, Journal of Alzheimers Disease, vol.43, pp.259-273, 2015.

M. E. López, A. Turrero, P. Cuesta, D. López-sanz, R. Bruña et al., Searching for Primary Predictors of Conversion from Mild Cognitive Impairment to Alzheimer's Disease: A Multivariate Follow-Up Study, J Alzheimers Dis, vol.52, pp.133-143, 2016.

W. Deeb, J. J. Giordano, P. J. Rossi, A. Y. Mogilner, A. Gunduz et al., Proceedings of the Fourth Annual Deep Brain Stimulation Think Tank: A Review of Emerging Issues and Technologies, Front Integr Neurosci, issue.10, 2016.

S. Bick and E. N. Eskandar, Neuromodulation for restoring memory, Neurosurg Focus, vol.40, p.5, 2016.

N. Suthana, Z. Haneef, J. Stern, R. Mukamel, E. Behnke et al., Memory Enhancement and Deep-Brain Stimulation of the Entorhinal Area, N Engl J Med, vol.366, pp.502-510, 2012.

J. Kuhn, K. Hardenacke, D. Lenartz, T. Gruendler, M. Ullsperger et al., Deep brain stimulation of the nucleus basalis of Meynert in Alzheimer's dementia, Mol Psychiatry, vol.20, pp.353-360, 2015.

A. W. Laxton, D. F. Tang-wai, M. P. Mcandrews, D. Zumsteg, R. Wennberg et al., A phase I trial of deep brain stimulation of memory circuits in Alzheimer's disease, Ann Neurol, vol.68, pp.521-534, 2010.

D. Fontaine, A. Deudon, J. J. Lemaire, J. Darcourt, P. Paquis et al., Traitement symptomatique des troubles mnésiques de la maladie d'Alzheimer par stimulation électrique chronique du fornix dans l'hypothalamus, Neurochirurgie, vol.58, p.411, 2012.

T. Sankar, M. M. Chakravarty, A. Bescos, M. Lara, T. Obuchi et al., Deep Brain Stimulation Influences Brain Structure in Alzheimer's Disease, Brain Stimulation, vol.8, pp.645-654, 2015.

F. A. Ponce, W. Asaad, K. D. Foote, W. S. Anderson, R. Cosgrove et al., 130 Bilateral Fornix Deep Brain Stimulation for Alzheimer Disease, Neurosurgery, vol.62, p.207, 2015.

G. S. Smith, A. W. Laxton, D. F. Tang-wai, M. P. Mcandrews, A. O. Diaconescu et al., Increased Cerebral Metabolism After 1 Year of Deep Brain Stimulation in Alzheimer Disease, Arch Neurol, p.69, 2012.

F. A. Ponce, W. F. Asaad, K. D. Foote, W. S. Anderson, R. Cosgrove et al., for The ARG. Bilateral deep brain stimulation of the fornix for Alzheimer's disease: surgical safety in the ADvance trial, J Neurosurg, vol.125, pp.75-84, 2016.

A. M. Lozano, L. Fosdick, M. M. Chakravarty, J. Leoutsakos, C. Munro et al., A phase II study of fornix deep brain stimulation in mild Alzheimer's disease, J Alzheimers Dis, vol.54, pp.777-787, 2016.

I. Gonsalvez, R. Baror, P. Fried, E. Santarnecchi, and A. Pascual-leone, Therapeutic noninvasive brain stimulation in Alzheimer's disease, Current Alzheimer research, vol.14, pp.362-376, 2017.

P. Caliandro, L. Padua, A. Rossi, P. M. Rossini, E. Stalberg et al., Jitter of Corticospinal Neurons During Repetitive Transcranial Magnetic Stimulation. Method and Possible Clinical Implications, Brain Stimulation, vol.7, pp.580-586, 2014.

T. Wagner, F. Fregni, S. Fecteau, A. Grodzinsky, M. Zahn et al., Transcranial direct current stimulation: A computer-based human model study, Neuroimage, vol.35, pp.1113-1124, 2007.

J. Lefaucheur, N. André-obadia, A. Antal, S. S. Ayache, C. Baeken et al., Evidence-based guidelines on the therapeutic use of repetitive transcranial magnetic stimulation (rTMS), Clin Neurophysiol, vol.125, pp.2150-2206, 2014.

J. Lefaucheur, A. Antal, S. S. Ayache, D. H. Benninger, J. Brunelin et al., Evidence-based guidelines on the therapeutic use of transcranial direct current stimulation (tDCS), Clin Neurophysiol, vol.128, pp.56-92, 2017.

M. C. Ridding and J. C. Rothwell, Is there a future for therapeutic use of transcranial magnetic stimulation?, Nature Reviews Neuroscience, vol.8, pp.559-567, 2007.

M. A. Ahmed, E. S. Darwish, E. M. Khedr, Y. M. El-serogy, and A. M. Ali, Effects of low versus high frequencies of repetitive transcranial magnetic stimulation on cognitive function and cortical excitability in Alzheimer's dementia, J Neurol, vol.259, pp.83-92, 2011.

J. M. Rabey, E. Dobronevsky, S. Aichenbaum, O. Gonen, R. G. Marton et al., Repetitive transcranial magnetic stimulation combined with cognitive training is a safe and effective modality for the treatment of Alzheimer's disease: a randomized, double-blind study, J Neural Transm (Vienna), vol.120, pp.813-819, 2013.

J. M. Rabey and E. Dobronevsky, Repetitive transcranial magnetic stimulation (rTMS) combined with cognitive training is a safe and effective modality for the treatment of Alzheimer's disease: clinical experience, J Neural Transm, vol.123, pp.1449-1455, 2016.

N. Toschi, M. E. Keck, T. Welt, and M. Guerrisi, Quantifying uncertainty in Transcranial Magnetic Stimulation-A high resolution simulation study in ICBM space, Conf Proc IEEE Eng Med Biol Soc, vol.2012, pp.1218-1221, 2012.

N. Toschi, T. Welt, M. Guerrisi, and M. E. Keck, Transcranial magnetic stimulation in heterogeneous brain tissue: clinical impact on focality, reproducibility and true sham stimulation, J Psychiatr Res, vol.43, pp.255-264, 2009.

N. Toschi, T. Welt, M. Guerrisi, and M. E. Keck, A reconstruction of the conductive phenomena elicited by transcranial magnetic stimulation in heterogeneous brain tissue, Phys Med, vol.24, pp.80-86, 2008.

E. Tatti, S. Rossi, I. Innocenti, A. Rossi, and E. Santarnecchi, Non-invasive brain stimulation of the aging brain: State of the art and future perspectives, Ageing Research Reviews, vol.29, pp.66-89, 2016.

R. Goutagny, N. Gu, C. Cavanagh, J. Jackson, J. Chabot et al., Alterations in hippocampal network oscillations and theta-gamma coupling arise before A? overproduction in a mouse model of Alzheimer's disease, Eur J Neurosci, vol.37, pp.1896-1902, 2013.

H. F. Iaccarino, A. C. Singer, A. J. Martorell, A. Rudenko, F. Gao et al., Gamma frequency entrainment attenuates amyloid load and modifies microglia, Nature, vol.540, pp.230-235, 2016.

C. S. Musaeus, M. M. Shafi, E. Santarnecchi, S. T. Herman, and D. Z. Press, Levetiracetam Alters Oscillatory Connectivity in Alzheimer's Disease, J Alzheimers Dis, vol.58, pp.1065-1076, 2017.

A. Antal, I. Alekseichuk, M. Bikson, J. Brockmöller, A. R. Brunoni et al., Low intensity transcranial electric stimulation: Safety, ethical, legal regulatory and application guidelines, Clin Neurophysiol, vol.128, pp.1774-1809, 2017.

F. Fröhlich and D. A. Mccormick, Endogenous Electric Fields May Guide Neocortical Network Activity, Neuron, vol.67, pp.129-143, 2010.

D. Reato, A. Rahman, M. Bikson, and L. C. Parra, Low-Intensity Electrical Stimulation Affects Network Dynamics by Modulating Population Rate and Spike Timing, J Neurosci, vol.30, pp.15067-15079, 2010.

M. Feurra, G. Bianco, D. Testa, M. Santarnecchi, E. Rossi et al., 21 Frequencydependent tuning of human motor system induced by transcranial oscillatory potentials, Clin Neurophysiol, vol.122, p.126, 2011.

M. Feurra, P. Pasqualetti, G. Bianco, E. Santarnecchi, A. Rossi et al., State-Dependent Effects of Transcranial Oscillatory Currents on the Motor System: What You Think Matters, J Neurosci, vol.33, pp.17483-17489, 2013.

E. Santarnecchi, P. Nicola, R. Godone, M. Giovannelli, F. Feurra et al., Frequency-Dependent Enhancement of Fluid Intelligence Induced by Transcranial Oscillatory Potentials, Curr Biol, vol.23, pp.1449-1453, 2013.

E. Santarnecchi, T. Muller, S. Rossi, A. Sarkar, N. R. Polizzotto et al., Individual differences and specificity of prefrontal gamma frequency-tACS on fluid intelligence capabilities, Cortex, vol.75, pp.33-43, 2016.

E. Santarnecchi, A. Biasella, E. Tatti, A. Rossi, D. Prattichizzo et al., High-gamma oscillations in the motor cortex during visuo-motor coordination: A tACS interferential study, Brain Res Bull, vol.131, pp.47-54, 2017.

C. R. Jack, H. J. Wiste, S. D. Weigand, T. M. Therneau, V. J. Lowe et al., Defining imaging biomarker cut points for brain aging and Alzheimer's disease, vol.13, pp.205-216, 2017.

N. L. Foster, J. L. Heidebrink, C. M. Clark, W. J. Jagust, S. E. Arnold et al., FDG-PET improves accuracy in distinguishing frontotemporal dementia and Alzheimer's disease, Brain, vol.130, pp.2616-2635, 2007.

M. Lehmann, P. M. Ghosh, C. Madison, R. Laforce, C. Corbetta-rastelli et al., Diverging patterns of amyloid deposition and hypometabolism in clinical variants of probable Alzheimer's disease, Brain, vol.136, pp.844-858, 2013.

N. M. Dowling, S. C. Johnson, C. E. Gleason, and W. J. Jagust, Initiative AsDN. The mediational effects of FDG hypometabolism on the association between cerebrospinal fluid biomarkers and neurocognitive function, Neuroimage, vol.105, pp.357-368, 2015.

S. Morbelli, R. Perneczky, A. Drzezga, G. B. Frisoni, A. Caroli et al., Metabolic Networks Underlying Cognitive Reserve in Prodromal Alzheimer Disease: A European Alzheimer Disease Consortium Project, J Nucl Med, vol.54, pp.894-902, 2013.

G. Chételat, R. Ossenkoppele, V. L. Villemagne, A. Perrotin, B. Landeau et al., Atrophy, hypometabolism and clinical trajectories in patients with amyloid-negative Alzheimer's disease, Brain, vol.139, pp.2528-2539, 2016.

R. H. Tan, J. J. Kril, Y. Yang, N. Tom, J. R. Hodges et al., Assessment of amyloid ? in pathologically confirmed frontotemporal dementia syndromes, Assessment & Disease Monitoring, vol.9, pp.10-20, 2017.

R. Ossenkoppele, W. J. Jansen, G. D. Rabinovici, D. L. Knol, W. M. Van-der-flier et al., Prevalence of Amyloid PET Positivity in Dementia Syndromes, vol.313, 1939.

J. R. Brosch, M. R. Farlow, S. L. Risacher, and L. G. Apostolova, Tau Imaging in Alzheimer's Disease Diagnosis and Clinical Trials, Neurotherapeutics, vol.14, pp.62-68, 2016.

B. Janssen, D. J. Vugts, U. Funke, G. T. Molenaar, P. S. Kruijer et al., Imaging of neuroinflammation in Alzheimer's disease, multiple sclerosis and stroke: Recent developments in positron emission tomography, Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, vol.1862, pp.425-441, 2016.

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 18 F-DPA-714 PET imaging, Brain, vol.139, pp.1252-1264, 2016.

K. Hiraoka, N. Okamura, Y. Funaki, A. Hayashi, M. Tashiro et al., Cholinergic Deficit and Response to Donepezil Therapy in Parkinson's Disease with Dementia, Eur Neurol, vol.68, pp.137-143, 2012.

M. Ziebell, B. B. Andersen, L. H. Pinborg, G. M. Knudsen, J. Stokholm et al., Striatal Dopamine Transporter Binding Does Not Correlate with Clinical Severity in Dementia with Lewy Bodies, J Nucl Med, vol.54, pp.1072-1076, 2013.

L. Chang, J. Lowe, A. Ardiles, J. Lim, A. C. Grey et al., Alzheimer's disease in the human eye. Clinical tests that identify ocular and visual information processing deficit as biomarkers, Alzheimer's & Dementia, vol.10, pp.251-261, 2014.

A. A. Sadun, M. Borchert, E. Devita, D. R. Hinton, and C. J. Bassi, Assessment of Visual Impairment in Patients With Alzheimer's Disease, Am J Ophthalmol, vol.104, pp.113-120, 1987.

G. C. Gilmore, H. E. Wenk, L. A. Naylor, and E. Koss, Motion Perception and Alzheimer's Disease, J Gerontol, vol.49, pp.52-57, 1994.

G. L. Trick, L. R. Trick, P. Morris, and M. Wolf, Visual field loss in senile dementia of the Alzheimer's type, Neurology, vol.45, pp.68-74, 1995.

S. L. Risacher, D. Wudunn, S. M. Pepin, T. R. Magee, B. C. Mcdonald et al., Visual contrast sensitivity in Alzheimer's disease, mild cognitive impairment, and older adults with cognitive complaints, Neurobiol Aging, vol.34, pp.1133-1144, 2013.

M. P. Bambo, E. Garcia-martin, S. Otin, J. Pinilla, J. M. Larrosa et al., Visual function and retinal nerve fibre layer degeneration in patients with Alzheimer disease: correlations with severity of dementia, Acta Ophthalmol, vol.93, pp.507-508, 2015.

R. Tzekov and M. Mullan, Vision function abnormalities in Alzheimer disease, Surv Ophthalmol, vol.59, pp.414-433, 2014.

Y. Ju, J. S. Mcleland, C. D. Toedebusch, C. Xiong, A. M. Fagan et al., Sleep Quality and Preclinical Alzheimer Disease, JAMA Neurology, vol.70, p.587, 2013.

L. Morgia, C. Ross-cisneros, F. N. Koronyo, Y. Hannibal, J. Gallassi et al., Melanopsin retinal ganglion cell loss in Alzheimer disease, Ann Neurol, vol.79, pp.90-109, 2016.

J. Chen, M. A. Espeland, R. L. Brunner, L. C. Lovato, R. B. Wallace et al., Sleep duration, cognitive decline, and dementia risk in older women, Alzheimer's & Dementia, vol.12, pp.21-33, 2016.

L. Chwiszczuk, M. Breitve, M. Hynninen, M. D. Gjerstad, D. Aarsland et al., Higher Frequency and Complexity of Sleep Disturbances in Dementia with Lewy Bodies as Compared to Alzheimer's Disease. Neurodegenerative Diseases, vol.16, pp.152-160, 2015.

F. Emamian, H. Khazaie, M. Tahmasian, G. D. Leschziner, M. J. Morrell et al., The Association Between Obstructive Sleep Apnea and Alzheimer's Disease: A Meta-Analysis Perspective, Front Aging Neurosci, issue.8, 2016.

H. Katsunuma, T. Shimizu, K. Ogawa, H. Kubo, H. Ishida et al., Treatment of insomnia by concomitant therapy with Zopiclone and Aniracetam in patients with cerebral infarction, cerebroatrophy, Alzheimer's disease and Parkinson's disease, Psychiatry Clin Neurosci, vol.52, pp.198-200, 1998.

M. V. Vitiello and S. Borson, Sleep disturbances in patients with Alzheimer's disease: epidemiology, pathophysiology and treatment, CNS Drugs, vol.15, pp.777-796, 2001.

N. J. Hart, Y. Koronyo, K. L. Black, and M. Koronyo-hamaoui, Ocular indicators of Alzheimer's: exploring disease in the retina, Acta Neuropathol, vol.132, pp.767-787, 2016.

R. Armstrong and H. Kergoat, Oculo-visual changes and clinical considerations affecting older patients with dementia, Ophthalmic Physiol Opt, vol.35, pp.352-376, 2015.

H. A. Bayhan, A. Bayhan, S. Celikbilek, A. Tanik, N. Gurdal et al., Evaluation of the chorioretinal thickness changes in Alzheimer's disease using spectral-domain optical coherence tomography, Clin Exp Ophthalmol, vol.43, pp.145-151, 2015.

J. C. Blanks, D. R. Hinton, A. A. Sadun, and C. A. Miller, Retinal ganglion cell degeneration in Alzheimer's disease, Brain Res, vol.501, pp.364-372, 1989.

J. C. Blanks, S. Y. Schmidt, Y. Torigoe, K. V. Porrello, D. R. Hinton et al., Retinal pathology in Alzheimer's disease. II. Regional neuron loss and glial changes in GCL, Neurobiol Aging, vol.17, pp.385-395, 1996.

G. Coppola, D. Renzo, A. Ziccardi, L. Martelli, F. Fadda et al., Optical Coherence Tomography in Alzheimer's Disease: A Meta-Analysis, PLoS One, vol.10, p.134750, 2015.

G. T. Feke, B. T. Hyman, R. A. Stern, and L. R. Pasquale, Retinal blood flow in mild cognitive impairment and Alzheimer's disease, Assessment & Disease Monitoring, vol.1, pp.144-151, 2015.

S. Frost, Y. Kanagasingam, H. Sohrabi, J. Vignarajan, P. Bourgeat et al., Retinal vascular biomarkers for early detection and monitoring of Alzheimer's disease, Translational Psychiatry, vol.3, p.233, 2013.

M. Gharbiya, A. Trebbastoni, F. Parisi, S. Manganiello, F. Cruciani et al., Choroidal thinning as a new finding in Alzheimer's disease: evidence from enhanced depth imaging spectral domain optical coherence tomography, J Alzheimers Dis, vol.40, pp.907-917, 2014.

L. E. Goldstein, J. A. Muffat, R. A. Cherny, R. D. Moir, M. H. Ericsson et al., Cytosolic ?-amyloid deposition and supranuclear cataracts in lenses from people with Alzheimer's disease. The Lancet, vol.361, pp.1258-1265, 2003.

D. R. Hinton, A. A. Sadun, J. C. Blanks, and C. A. Miller, Optic-Nerve Degeneration in Alzheimer's Disease, N Engl J Med, vol.315, pp.485-487, 1986.

H. Kergoat, M. Kergoat, L. Justino, H. Chertkow, A. Robillard et al., An evaluation of the retinal nerve fiber layer thickness by scanning laser polarimetry in individuals with dementia of the Alzheimer type, Acta Ophthalmol Scand, vol.79, pp.187-191, 2001.

S. Kirbas, K. Turkyilmaz, O. Anlar, A. Tufekci, and M. Durmus, Retinal Nerve Fiber Layer Thickness in Patients With Alzheimer Disease, J Neuroophthalmol, vol.33, pp.58-61, 2013.

Y. Koronyo, D. Biggs, E. Barron, D. S. Boyer, J. A. Pearlman et al., Retinal amyloid pathology and proof-of-concept imaging trial in Alzheimer's disease, JCI Insight, issue.2, 2017.

C. Paquet, M. Boissonnot, F. Roger, P. Dighiero, R. Gil et al., Abnormal retinal thickness in patients with mild cognitive impairment and Alzheimer's disease, Neurosci Lett, vol.420, pp.97-99, 2007.

N. Patton, T. Aslam, T. Macgillivray, P. A. Deary, I. J. Dhillon et al., Retinal vascular image analysis as a potential screening tool for cerebrovascular disease: a rationale based on homology between cerebral and retinal microvasculatures, J Anat, vol.206, pp.319-348, 2005.

A. A. Sadun and C. J. Bassi, Optic Nerve Damage in Alzheimer's Disease, Ophthalmology, vol.97, pp.9-17, 1990.

C. Schön, N. A. Hoffmann, S. M. Ochs, S. Burgold, S. Filser et al., Long-Term In Vivo Imaging of Fibrillar Tau in the Retina of P301S Transgenic Mice, PLoS One, vol.7, p.53547, 2012.

M. Koronyo-hamaoui, Y. Koronyo, A. V. Ljubimov, C. A. Miller, M. K. Ko et al., Identification of amyloid plaques in retinas from Alzheimer's patients and noninvasive in vivo optical imaging of retinal plaques in a mouse model, Neuroimage, vol.54, pp.204-217, 2011.

F. Berisha, G. T. Feke, C. L. Trempe, J. W. Mcmeel, and C. L. Schepens, Retinal Abnormalities in Early Alzheimer's Disease. Investigative Opthalmology & Visual Science, vol.48, p.2285, 2007.

C. Y. Cheung, Y. T. Ong, M. K. Ikram, S. Y. Ong, X. Li et al., Microvascular network alterations in the retina of patients with Alzheimer's disease, Alzheimer's & Dementia, vol.10, pp.135-142, 2014.

C. A. Curcio and D. N. Drucker, Retinal ganglion cells in Alzheimer's disease and aging, Ann Neurol, vol.33, pp.248-257, 2004.

S. Frost, R. N. Martins, and Y. Kanagasingam, Ocular Biomarkers for Early Detection of Alzheimer's Disease, J Alzheimers Dis, vol.22, pp.1-16, 2010.

L. Gao, Y. Liu, X. Li, Q. Bai, and P. Liu, Abnormal retinal nerve fiber layer thickness and macula lutea in patients with mild cognitive impairment and Alzheimer's disease, Arch Gerontol Geriatr, vol.60, pp.162-167, 2015.

L. Guo, J. Duggan, and M. Cordeiro, Alzheimers Disease and Retinal Neurodegeneration, Current Alzheimer Research, vol.7, pp.3-14, 2010.

A. Kapasi and J. A. Schneider, Vascular contributions to cognitive impairment, clinical Alzheimer's disease, and dementia in older persons, Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, vol.1862, pp.878-886, 2016.

R. Kromer, N. Serbecic, L. Hausner, L. Froelich, F. Aboul-enein et al., Detection of Retinal Nerve Fiber Layer Defects in Alzheimer's Disease Using SD-OCT, Frontiers in Psychiatry, issue.5, 2014.

D. Liu, L. Zhang, Z. Li, X. Zhang, Y. Wu et al., Thinner changes of the retinal nerve fiber layer in patients with mild cognitive impairment and Alzheimer's disease, BMC Neurol, p.15, 2015.

E. Marziani, S. Pomati, P. Ramolfo, M. Cigada, A. Giani et al., Evaluation of Retinal Nerve Fiber Layer and Ganglion Cell Layer Thickness in Alzheimer's Disease Using Spectral-Domain Optical Coherence Tomography, Investigative Opthalmology & Visual Science, vol.54, p.5953, 2013.

T. Moreno-ramos, J. Benito-leon, A. Villarejo, and F. Bermejo-pareja, Retinal Nerve Fiber Layer Thinning in Dementia Associated with Parkinson's Disease, Dementia with Lewy Bodies, and Alzheimer's Disease, Journal of Alzheimers Disease, vol.34, pp.659-664, 2013.

M. M. Moschos, I. Markopoulos, I. Chatziralli, A. Rouvas, S. G. Papageorgiou et al., Structural and Functional Impairment of the Retina and Optic Nerve in Alzheimer's Disease, Current Alzheimer Research, vol.9, pp.782-788, 2012.

J. M. Nolan, E. Loskutova, A. N. Howard, R. Moran, R. Mulcahy et al., Macular Pigment, Visual Function, and Macular Disease among Subjects with Alzheimer's Disease: An Exploratory Study, Journal of Alzheimers Disease, vol.42, pp.1191-1202, 2014.

V. Parisi, R. Restuccia, F. Fattapposta, C. Mina, M. G. Bucci et al., Morphological and functional retinal impairment in Alzheimer's disease patients, Clin Neurophysiol, vol.112, pp.1860-1867, 2001.

Z. Y. Shi, Y. J. Wu, M. J. Wang, J. Cao, W. Feng et al., Greater Attenuation of Retinal Nerve Fiber Layer Thickness in Alzheimer's Disease Patients, Journal of Alzheimers Disease, vol.40, pp.277-283, 2014.

G. L. Trick, M. C. Barris, and M. Bickler-bluth, Abnormal pattern electroretinograms in patients with senile dementia of the alzheimer type, Ann Neurol, vol.26, pp.226-231, 1989.

M. A. Williams, A. J. Mcgowan, C. R. Cardwell, C. Y. Cheung, D. Craig et al.,

, Alzheimer's & Dementia: Diagnosis, Assessment & Disease Monitoring, vol.1, pp.229-235, 2015.

D. Purves, G. J. Augustine, D. Fitzpatrick, L. C. Katz, A. Lamantia et al., The Retina. Sinauer Associates

(. Sunderland and . Ma, , 2001.

M. S. Byerly and S. Blackshaw, Vertebrate retina and hypothalamus development, Wiley Interdiscip Rev Syst Biol Med, vol.1, pp.380-389, 2009.

A. Trost, S. Lange, F. Schroedl, D. Bruckner, K. A. Motloch et al., Brain and Retinal Pericytes: Origin, Function and Role, Front Cell Neurosci, issue.10, 2016.

E. Vecino, F. D. Rodriguez, N. Ruzafa, X. Pereiro, and S. C. Sharma, Glia-neuron interactions in the mammalian retina, Prog Retin Eye Res, vol.51, pp.1-40, 2016.

P. J. Morin, C. R. Abraham, A. Amaratunga, R. J. Johnson, G. Huber et al., Amyloid Precursor Protein Is Synthesized by Retinal Ganglion Cells, Rapidly Transported to the Optic Nerve Plasma Membrane and Nerve Terminals, and Metabolized, J Neurochem, vol.61, pp.464-473, 2006.

J. Cai, X. Qi, N. Kociok, S. Skosyrski, A. Emilio et al., Boulton ME. ?-Secretase (BACE1) inhibition causes retinal pathology by vascular dysregulation and accumulation of age pigment, EMBO Mol Med, vol.4, pp.980-991, 2012.

L. Li, J. Luo, D. Chen, J. B. Tong, L. P. Zeng et al., BACE1 in the retina: a sensitive biomarker for monitoring early pathological changes in Alzheimer's disease, Neural Regeneration Research, vol.11, pp.447-453, 2016.

P. N. Alexandrov, A. Pogue, S. Bhattacharjee, and W. J. Lukiw, Retinal amyloid peptides and complement factor H in transgenic models of Alzheimer's disease, Neuroreport, vol.22, pp.623-627, 2011.

Y. Tsai, B. Lu, A. V. Ljubimov, S. Girman, F. N. Ross-cisneros et al., Ocular Changes in TgF344-AD Rat Model of Alzheimer's Disease, Investigative Opthalmology & Visual Science, vol.55, p.523, 2014.

C. Y. Ho, J. C. Troncoso, D. Knox, W. Stark, and C. G. Eberhart, Beta-Amyloid, Phospho-Tau and AlphaSynuclein Deposits Similar to Those in the Brain Are Not Identified in the Eyes of Alzheimer's and Parkinson's Disease Patients, Brain Pathol, vol.24, pp.25-32, 2014.

A. Kesler, V. Vakhapova, A. D. Korczyn, E. Naftaliev, and M. Neudorfer, Retinal thickness in patients with mild cognitive impairment and Alzheimer's disease, Clin Neurol Neurosurg, vol.113, pp.523-526, 2011.

Y. Lu, Z. Li, X. Zhang, B. Ming, J. Jia et al., Retinal nerve fiber layer structure abnormalities in early Alzheimer's disease: Evidence in optical coherence tomography, Neurosci Lett, vol.480, pp.69-72, 2010.

M. F. Cordeiro, L. Guo, V. Luong, G. Harding, W. Wang et al., Real-time imaging of single nerve cell apoptosis in retinal neurodegeneration, Proceedings of the National Academy of Sciences, vol.101, pp.13352-13356, 2004.

L. Y. Du, L. Chang, A. O. Ardiles, C. Tapia-rojas, J. Araya et al., Alzheimer's Disease-Related Protein Expression in the Retina of Octodon degus, PLoS One, vol.10, p.135499, 2015.

R. M. Dutescu, Q. Li, J. Crowston, C. L. Masters, P. N. Baird et al., Amyloid precursor protein processing and retinal pathology in mouse models of Alzheimer's disease. Graefe's Archive for, Clinical and Experimental Ophthalmology, vol.247, pp.1213-1221, 2009.

M. M. Edwards, J. J. Rodríguez, R. Gutierrez-lanza, J. Yates, A. Verkhratsky et al., Retinal macroglia changes in a triple transgenic mouse model of Alzheimer's disease, Exp Eye Res, vol.127, pp.252-260, 2014.

L. Gao, X. Chen, Y. Tang, J. Zhao, Q. Li et al., Neuroprotective effect of memantine on the retinal ganglion cells of APPswe/PS1?E9 mice and its immunomodulatory mechanisms, Exp Eye Res, vol.135, pp.47-58, 2015.

V. K. Gupta, N. Chitranshi, V. B. Gupta, M. Golzan, Y. Dheer et al., Amyloid ? accumulation and inner retinal degenerative changes in Alzheimer's disease transgenic mouse, Neurosci Lett, vol.623, pp.52-56, 2016.

Y. He, H. Zhao, and G. Su, Ginsenoside Rg1 Decreases Neurofibrillary Tangles Accumulation in Retina by Regulating Activities of Neprilysin and PKA in Retinal Cells of AD Mice Model, J Mol Neurosci, vol.52, pp.101-106, 2013.

Y. Koronyo, B. C. Salumbides, K. L. Black, and M. Koronyo-hamaoui, Alzheimer's Disease in the Retina: Imaging Retinal A? Plaques for Early Diagnosis and Therapy Assessment, Neurodegenerative Diseases, vol.10, pp.285-293, 2012.

B. Liu, S. Rasool, Z. Yang, C. G. Glabe, S. S. Schreiber et al., Amyloid-peptide vaccinations reduce {beta}-amyloid plaques but exacerbate vascular deposition and inflammation in the retina of Alzheimer's transgenic mice, Am J Pathol, vol.175, pp.2099-2110, 2009.

S. S. More and R. Vince, Hyperspectral Imaging Signatures Detect Amyloidopathy in Alzheimer's Mouse Retina Well before Onset of Cognitive Decline, ACS Chem Neurosci, vol.6, pp.306-315, 2015.

A. Ning, J. Cui, E. To, K. H. Ashe, and J. Matsubara, Amyloid-? Deposits Lead to Retinal Degeneration in a Mouse Model of Alzheimer Disease, Investigative Opthalmology & Visual Science, vol.49, p.5136, 2008.

S. W. Park, J. H. Kim, I. Mook-jung, K. Kim, W. J. Park et al., Intracellular amyloid beta alters the tight junction of retinal pigment epithelium in 5XFAD mice, Neurobiol Aging, vol.35, pp.2013-2020, 2014.

R. Parthasarathy, K. M. Chow, Z. Derafshi, M. P. Fautsch, J. R. Hetling et al., Reduction of amyloid-beta levels in mouse eye tissues by intra-vitreally delivered neprilysin, Exp Eye Res, vol.138, pp.134-144, 2015.

S. E. Perez, S. Lumayag, B. Kovacs, E. J. Mufson, and S. Xu, ?-Amyloid Deposition and Functional Impairment in the Retina of the APPswe/PS1?E9 Transgenic Mouse Model of Alzheimer's Disease, Investigative Opthalmology & Visual Science, vol.50, p.793, 2009.

A. I. Pogue, P. Dua, J. M. Hill, and W. J. Lukiw, Progressive inflammatory pathology in the retina of aluminum-fed 5xFAD transgenic mice, J Inorg Biochem, vol.152, pp.206-209, 2015.

P. A. Williams, R. A. Thirgood, H. Oliphant, A. Frizzati, E. Littlewood et al., Retinal ganglion cell dendritic degeneration in a mouse model of Alzheimer's disease, Neurobiol Aging, vol.34, pp.1799-1806, 2013.

Y. Yang, C. Shiao, J. F. Hemingway, N. L. Jorstad, B. R. Shalloway et al., Suppressed Retinal Degeneration in Aged Wild Type and APPswe/PS1?E9 Mice by Bone Marrow Transplantation, PLoS One, vol.8, 2013.

H. Zhao, R. Chang, C. H. Wang, J. Yang, L. Fang et al., Hyperphosphorylation of tau protein by calpain regulation in retina of Alzheimer's disease transgenic mouse, Neurosci Lett, vol.551, pp.12-16, 2013.

A. N. Begum, M. R. Jones, G. P. Lim, T. Morihara, P. Kim et al., Curcumin Structure-Function, Bioavailability, and Efficacy in Models of Neuroinflammation and Alzheimer's Disease, J Pharmacol Exp Ther, vol.326, pp.196-208, 2008.

P. Dadhaniya, C. Patel, J. Muchhara, N. Bhadja, N. Mathuria et al., Safety assessment of a solid lipid curcumin particle preparation: Acute and subchronic toxicity studies, Food Chem Toxicol, vol.49, pp.1834-1842, 2011.

M. Garcia-alloza, L. A. Borrelli, A. Rozkalne, B. T. Hyman, and B. J. Bacskai, Curcumin labels amyloid pathologyin vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model, J Neurochem, vol.102, pp.1095-1104, 2007.

K. G. Goozee, T. M. Shah, H. R. Sohrabi, S. R. Rainey-smith, B. Brown et al., Examining the potential clinical value of curcumin in the prevention and diagnosis of Alzheimer's disease, Br J Nutr, vol.115, pp.449-465, 2015.

V. S. Gota, G. B. Maru, T. G. Soni, T. R. Gandhi, N. Kochar et al., Safety and Pharmacokinetics of a Solid Lipid Curcumin Particle Formulation in Osteosarcoma Patients and Healthy Volunteers, J Agric Food Chem, vol.58, pp.2095-2099, 2010.

P. Kumaraswamy, S. Sethuraman, and U. M. Krishnan, Mechanistic Insights of Curcumin Interactions with the Core-Recognition Motif of ?-Amyloid Peptide, J Agric Food Chem, vol.61, pp.3278-3285, 2013.

Y. Masuda, M. Fukuchi, T. Yatagawa, M. Tada, K. Takeda et al., Solid-state NMR analysis of interaction sites of curcumin and 42-residue amyloid ?-protein fibrils, Bioorg Med Chem, vol.19, pp.5967-5974, 2011.

M. Mutsuga, J. K. Chambers, K. Uchida, M. Tei, T. Makibuchi et al., Binding of Curcumin to Senile Plaques and Cerebral Amyloid Angiopathy in the Aged Brain of Various Animals and to Neurofibrillary Tangles in Alzheimer's Brain, J Vet Med Sci, vol.74, pp.51-57, 2012.

F. Yang, G. P. Lim, A. N. Begum, O. J. Ubeda, M. R. Simmons et al., Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo, J Biol Chem, vol.280, pp.5892-5901, 2005.

E. K. Ryu, Y. S. Choe, K. Lee, Y. Choi, and B. Kim, Curcumin and Dehydrozingerone Derivatives: Synthesis, Radiolabeling, and Evaluation for ?-Amyloid Plaque Imaging ?, J Med Chem, vol.49, pp.6111-6119, 2006.

M. Tei, K. Uchida, M. Mutsuga, J. K. Chambers, and H. Nakayama, The Binding of Curcumin to Various Types of Canine Amyloid Proteins, J Vet Med Sci, vol.74, pp.481-483, 2012.

D. Yanagisawa, T. Amatsubo, S. Morikawa, H. Taguchi, M. Urushitani et al., In vivo detection of amyloid ? deposition using 19F magnetic resonance imaging with a 19F-containing curcumin derivative in a mouse model of Alzheimer's disease, Neuroscience, vol.184, pp.120-127, 2011.

D. Yanagisawa, N. Shirai, T. Amatsubo, H. Taguchi, K. Hirao et al., Relationship between the tautomeric structures of curcumin derivatives and their A?-binding activities in the context of therapies for Alzheimer's disease, Biomaterials, vol.31, pp.4179-4185, 2010.

D. Yanagisawa, H. Taguchi, A. Yamamoto, N. Shirai, K. Hirao et al., Curcumin has binding activity to ?-amyloid oligomer, Alzheimer's & Dementia, vol.7, pp.476-477, 2011.

O. Butovsky, M. Koronyo-hamaoui, G. Kunis, E. Ophir, G. Landa et al., Glatiramer acetate fights against Alzheimer's disease by inducing dendritic-like microglia expressing insulin-like growth factor 1, Proceedings of the National Academy of Sciences, vol.103, pp.11784-11789, 2006.

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, vol.138, pp.2399-2422, 2015.

U. Kayabasi, Retinal Examination for the Diagnosis of Alzheimer's Disease, International Journal of Ophthalmic & Pathology, p.3, 2014.

P. J. Snyder, L. N. Johnson, Y. Y. Lim, C. Y. Santos, J. Alber et al., Nonvascular retinal imaging markers of preclinical Alzheimer's disease, Assessment & Disease Monitoring, vol.4, pp.169-178, 2016.

A. B. Einarsdottir, S. H. Hardarson, J. V. Kristjansdottir, D. T. Bragason, J. Snaedal et al., Retinal Oximetry Imaging in Alzheimer's Disease, J Alzheimers Dis, vol.49, pp.79-83, 2015.

E. Stefánsson, O. B. Olafsdottir, A. B. Einarsdottir, T. S. Eliasdottir, T. Eysteinsson et al., Retinal Oximetry Discovers Novel Biomarkers in Retinal and Brain Diseases, Investigative Opthalmology & Visual Science, vol.58, p.227, 2017.

N. K. Archibald, M. P. Clarke, U. P. Mosimann, and D. J. Burn, The retina in Parkinson's disease, Brain, vol.132, pp.1128-1145, 2009.

I. Bodis-wollner, P. B. Kozlowski, S. Glazman, and S. Miri, ?-synuclein in the inner retina in parkinson disease, Ann Neurol, vol.75, pp.964-966, 2014.

P. A. Calabresi, L. J. Balcer, and E. M. Frohman, Retinal pathology in multiple sclerosis: insight into the mechanisms of neuronal pathology, Brain, vol.133, pp.1575-1577, 2010.

Y. T. Ong, D. Silva, D. A. Cheung, C. Y. Chang, H. M. Chen et al., Microvascular Structure and Network in the Retina of Patients With Ischemic Stroke, Stroke, vol.44, pp.2121-2127, 2013.

L. F. Scinto, K. R. Daffner, D. Dressler, B. I. Ransil, D. Rentz et al., A potential noninvasive neurobiological test for Alzheimer's disease. SCIENCE-NEW YORK THEN WASHINGTON, pp.1051-1051, 1994.

L. Scinto, C. Wu, K. Firla, K. Daffner, D. Saroff et al., Focal pathology in the Edinger-Westphal nucleus explains pupillary hypersensitivity in Alzheimer's disease, Acta Neuropathol, vol.97, pp.557-564, 1999.

W. Nowak, M. Pieniazek, M. Nakayama, and A. Hachol, International Conference on; IEEE, Frontiers of Signal Processing, pp.58-62, 2016.

E. Granholm, S. Morris, D. Galasko, C. Shults, R. E. Vukov et al., Tropicamide effects on pupil size and pupillary light reflexes in Alzheimer's and Parkinson's disease, Int J Psychophysiol, vol.47, pp.95-115, 2003.

S. Durrleman, X. Pennec, A. Trouvé, and N. Ayache, Statistical models of sets of curves and surfaces based on currents, Med Image Anal, vol.13, pp.793-808, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00816051

S. Durrleman, M. Prastawa, N. Charon, J. R. Korenberg, S. Joshi et al., Morphometry of anatomical shape complexes with dense deformations and sparse parameters, Neuroimage, vol.101, pp.35-49, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01015771

P. Gori, O. Colliot, L. Marrakchi-kacem, Y. Worbe, A. Routier et al., Lecture Notes in Computer Science, pp.275-287, 2015.

P. Gori, O. Colliot, L. Marrakchi-kacem, Y. Worbe, C. Poupon et al., A Bayesian framework for joint morphometry of surface and curve meshes in multi-object complexes, Med Image Anal, vol.35, pp.458-474, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01359423

J. Schiratti, S. Allassonniere, O. Colliot, and S. Durrleman, Adv Neural Inf Process Syst, pp.2404-2412, 2015.

I. Koval, J. B. Schiratti, A. Routier, M. Bacci, O. Colliot et al., Medical Image Computing and Computer Assisted Intervention-MICCAI 2017, pp.451-459, 2017.

S. Allassonnière, S. Durrleman, and E. Kuhn, Bayesian Mixed Effect Atlas Estimation with a Diffeomorphic Deformation Model, SIAM Journal on Imaging Sciences, vol.8, pp.1367-1395, 2015.

S. Durrleman, X. Pennec, A. Trouvé, G. Gerig, and N. Ayache, Medical Image Computing and Computer-Assisted Intervention-MICCAI, pp.297-304, 2009.

S. Durrleman, X. Pennec, A. Trouve, J. Braga, G. Gerig et al., Toward a comprehensive framework for the spatiotemporal statistical analysis of longitudinal shape data, Int J Comput Vis, vol.103, pp.22-59, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00813825

J. Fishbaugh, M. Prastawa, S. Durrleman, J. Piven, and G. Gerig, Medical Image Computing and Computer-Assisted Intervention-MICCAI, pp.731-738, 2012.

J. Fishbaugh, M. Prastawa, G. Gerig, and S. Durrleman, Lecture Notes in Computer Science, pp.718-729, 2013.

J. B. Schiratti, S. Allassonnière, O. Colliot, and S. Durrleman, A Bayesian mixed-effects model to learn trajectories of changes from repeated manifold-valued observations, J Mach Learn Res, vol.3, pp.1-48, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01540367

J. L. Cummings, T. Morstorf, and K. Zhong, Alzheimer's disease drug-development pipeline: few candidates, frequent failures, Alzheimers Res Ther, vol.6, p.37, 2014.

J. Cummings, T. Morstorf, and G. Lee, Alzheimer's drug-development pipeline, Translational Research & Clinical Interventions, vol.2, pp.222-232, 2016.

S. I. Berger and R. Iyengar, Network analyses in systems pharmacology, Bioinformatics, vol.25, pp.2466-2472, 2009.

M. Danhof, Systems pharmacology-Towards the modeling of network interactions, Eur J Pharm Sci, vol.94, pp.4-14, 2016.

M. Rask-andersen, M. S. Almén, and H. B. Schiöth, Trends in the exploitation of novel drug targets, Nature Reviews Drug Discovery, vol.10, pp.579-590, 2011.

A. L. Hopkins, Network pharmacology: the next paradigm in drug discovery, Nat Chem Biol, vol.4, pp.682-690, 2008.

M. Campillos, M. Kuhn, A. C. Gavin, L. J. Jensen, and P. Bork, Drug Target Identification Using SideEffect Similarity, Science, vol.321, pp.263-266, 2008.

L. Xie, X. Ge, H. Tan, L. Xie, Y. Zhang et al., Towards Structural Systems Pharmacology to Study Complex Diseases and Personalized Medicine, PLoS Comput Biol, vol.10, 2014.

K. Nikolic, L. Mavridis, T. Djikic, J. Vucicevic, D. Agbaba et al., Drug Design for CNS Diseases: Polypharmacological Profiling of Compounds Using Cheminformatic, 3DQSAR and Virtual Screening Methodologies, Front Neurosci, issue.10, 2016.

M. Shahid, S. Cheema, M. Klenner, A. Younesi, E. Hofmann-apitius et al., SVM Based Descriptor Selection and Classification of Neurodegenerative Disease Drugs for Pharmacological Modeling, Molecular Informatics, vol.32, pp.241-249, 2013.

N. T. Issa, J. Kruger, H. Wathieu, R. Raja, S. W. Byers et al., DrugGenEx-Net: a novel computational platform for systems pharmacology and gene expression-based drug repurposing, BMC Bioinformatics, p.17, 2016.

K. A. Fujita, M. Ostaszewski, Y. Matsuoka, S. Ghosh, E. Glaab et al., Integrating Pathways of Parkinson's Disease in a Molecular Interaction Map, Mol Neurobiol, vol.49, pp.88-102, 2013.

S. Mizuno, R. Iijima, S. Ogishima, M. Kikuchi, Y. Matsuoka et al., AlzPathway: a comprehensive map of signaling pathways of Alzheimer's disease, BMC Syst Biol, vol.6, p.52, 2012.

A. Iyappan, M. Gündel, M. Shahid, J. Wang, H. Li et al., Towards a Pathway Inventory of the Human Brain for Modeling Disease Mechanisms Underlying Neurodegeneration, J Alzheimers Dis, vol.52, pp.1343-1360, 2016.

S. G. Mueller, M. W. Weiner, L. J. Thal, R. C. Petersen, C. Jack et al., The Alzheimer's Disease Neuroimaging Initiative, Neuroimaging Clin N Am, vol.15, pp.869-877, 2005.

A. Malhotra, E. Younesi, M. Gündel, B. Müller, M. T. Heneka et al., ADO: A disease ontology representing the domain knowledge specific to Alzheimer's disease, Alzheimer's & Dementia, vol.10, pp.238-246, 2014.

A. T. Kodamullil, E. Younesi, M. Naz, S. Bagewadi, and M. Hofmann-apitius, Computable cause-andeffect models of healthy and Alzheimer's disease states and their mechanistic differential analysis, Alzheimer's & Dementia, vol.11, pp.1329-1339, 2015.

M. Emon, A. T. Kodamullil, R. Karki, E. Younesi, and M. Hofmann-apitius, Using Drugs as Molecular Probes: A Computational Chemical Biology Approach in Neurodegenerative Diseases, J Alzheimers Dis, vol.56, pp.677-686, 2017.

H. Geerts, P. A. Dacks, V. Devanarayan, M. Haas, Z. S. Khachaturian et al., Big data to smart data in Alzheimer's disease: The brain health modeling initiative to foster actionable knowledge, Alzheimer's & Dementia, vol.12, pp.1014-1021, 2016.

W. E. Balch, R. I. Morimoto, A. Dillin, and J. W. Kelly, Adapting proteostasis for disease intervention, Science, vol.319, pp.916-919, 2008.

J. Lim and Z. Y. Yue, Neuronal Aggregates: Formation, Clearance, and Spreading, Dev Cell, vol.32, pp.491-501, 2015.

G. Calandra-buonaura, N. Toschi, F. Provini, I. Corazza, F. Bisulli et al., Physiologic autonomic arousal heralds motor manifestations of seizures in nocturnal frontal lobe epilepsy: Implications for pathophysiology, Sleep Med, vol.13, pp.252-262, 2012.

G. Valenza, S. Orsolini, S. Diciotti, L. Citi, E. P. Scilingo et al., Assessment of spontaneous cardiovascular oscillations in Parkinson's disease, Biomedical Signal Processing and Control, vol.26, pp.80-89, 2016.

G. Valenza, L. Citi, R. G. Garcia, J. N. Taylor, N. Toschi et al., Complexity Variability Assessment of Nonlinear Time-Varying Cardiovascular Control, Sci Rep, vol.7, p.42779, 2017.

A. Duggento, M. Bianciardi, L. Passamonti, L. L. Wald, M. Guerrisi et al., Globally conditioned Granger causality in brain-brain and brain-heart interactions: a combined heart rate variability/ultra-high-field (7 T) functional magnetic resonance imaging study, Philos Trans A Math Phys Eng Sci, p.374, 2016.

N. Toschi, A. Duggento, A. Canichella, F. Coniglione, M. Dauri et al., Intra-and inter-beat modeling of cardiovascular dynamics and control: assessing haemodynamic stability and responsiveness, Conf Proc IEEE Eng Med Biol Soc, pp.8440-8443, 2011.

A. Duggento, N. Toschi, and M. Guerrisi, Modeling of Human Baroreflex: Considerations on the Seidel-Herzel Model. Fluctuation and Noise Letters, vol.11, p.1240017, 2012.

R. A. Poldrack, Precision Neuroscience: Dense Sampling of Individual Brains, Neuron, vol.95, pp.727-729, 2017.

E. M. Gordon, T. O. Laumann, A. W. Gilmore, D. J. Newbold, D. J. Greene et al., Precision Functional Mapping of Individual Human Brains, Neuron, vol.95, p.791, 2017.

R. M. Braga and R. L. Buckner, Parallel Interdigitated Distributed Networks within the Individual Estimated by Intrinsic Functional Connectivity, Neuron, vol.95, p.457, 2017.

B. Dubois, H. Hampel, H. H. Feldman, P. Scheltens, P. Aisen et al., Preclinical Alzheimer's disease: definition, natural history, and diagnostic criteria, vol.12, pp.292-323, 2016.

S. E. Petersen and O. Sporns, Brain networks and cognitive architectures, Neuron, vol.88, pp.207-219, 2015.

, CONTRIBUTORS TO THE ALZHEIMER PRECISION MEDICINE INITIATIVEWORKING GROUP

, Principal Investigator and Speaker

L. Aguilar, ). Montréal, C. Babiloni, ). Rome, F. Baldacci et al., Chiesa PA (Par-is), Colliot O (Paris), Coman CM (Paris), Graziani M (Roma), Haberkamp M (Bonn), Habert MO (Paris), Hampel H (Paris), Herholz K (Manches-ter), Karran E (Cambridge)

E. Santarnecchi, ). Siena, and L. S. Schneider, Sporns O (Bloomington), Toschi N (Rome), Verdooner SR (Sacramento), Vergallo A (Paris), Villain N (Paris)