B. Kastler and D. Vetter, Comprendre l'IRM: manuel d'auto-apprentissage, 2018.

P. Eustache, Evaluation par IRM multimodale des modifications cérébrales chez des patients Alzheimer à un stade prodromique. Optimisation de la relaxométrie T2* par IRM, 2015.

S. Mori and J. Zhang, Principles of Diffusion Tensor Imaging and Its Applications to Basic Neuroscience Research, Neuron, vol.51, issue.5, pp.527-539, 2006.

D. W. Mcrobbie, E. A. Moore, M. J. Graves, and M. R. Prince, Acronyms Anonymous I: Spin Echo, MRI from Picture to Proton, pp.185-206, 2017.

E. O. Stejskal and J. E. Tanner, Spin Diffusion Measurements: Spin Echoes in the Presence of a Time-Dependent Field Gradient, J. Chem. Phys, vol.42, issue.1, pp.288-292, 1965.

A. D. Courtesy-of and . Elster, MRIquestions.com, 2019.

T. Arthur, Brain Mapping an Encyclopedic Reference, 2015.

S. Mori, Three dimensional tracking axpma; [rpkectopms om tje braom by MRI, Ann. Neurol, vol.45, issue.2, pp.247-50, 1999.

D. S. Tuch, Q-ball imaging, Magn. Reson. Med, vol.52, issue.6, pp.1358-1372, 2004.

N. J. Tustison and J. C. Gee, Introducing Dice, Jaccard, and Other Label Overlap Measures To ITK, Insight J, issue.2, 2009.

H. Daniel, R. William, and K. Gregory, Comparing Images Using the Hausdorff Distance Under Translation, 1992.

A. W. Toga and P. M. Thompson, The role of image registration in brain mapping, Image Vis. Comput, vol.19, issue.1-2, pp.3-24, 2001.

D. J. Burr, Elastic matching of line drawings, IEEE Trans. Pattern Anal. Mach. Intell, issue.6, pp.708-713, 1981.

D. W. Eggert, A. Lorusso, and R. B. Fisher, Estimating 3-D rigid body transformations: a comparison of four major algorithms, 1997.

C. Studholme, D. L. Hill, and D. J. Hawkes, An overlap invariant entropy measure of 3D medical image alignment, Pattern Recognit, vol.32, issue.1, pp.71-86, 1999.

F. Maes, A. Collignon, D. Vandermeulen, G. Marchal, and P. Suetens, Multimodality image registration by maximization of mutual information, IEEE Trans. Med. Imaging, vol.16, issue.2, pp.187-198, 1997.

R. Shams, P. Sadeghi, R. Kennedy, and R. Hartley, Parallel computation of mutual information on the GPU with application to real-time registration of 3D medical images, Comput. Methods Programs Biomed, vol.99, issue.2, pp.133-146

A. Eklund, M. Andersson, and H. Knutsson, Linköping University Post Print Phase Based Volume Registration Using CUDA PHASE BASED VOLUME REGISTRATION USING CUDA, pp.658-661, 2010.

O. Fluck, C. Vetter, W. Wein, A. Kamen, B. Preim et al., A survey of medical image registration on graphics hardware, Comput. Methods Programs Biomed, vol.104, pp.45-57, 2010.

K. Punithakumar, P. Boulanger, and M. Noga, A GPU-Accelerated Deformable Image Registration Algorithm with Applications to Right Ventricular Segmentation, 2017.

C. J. Holmes, R. Hoge, L. Collins, R. Woods, A. W. Toga et al., Enhancement of MR Images Using Registration for Signal Averaging, J. Comput. Assist. Tomogr, vol.22, issue.2, pp.324-333, 1998.

M. Gierthmuehlen, Mapping of Sheep Sensory Cortex With a Novel Microelectrocorticography Grid, J. Comp. Neurol, vol.522, pp.3590-3608, 2014.

M. Fang, J. Li, J. A. Rudd, S. M. Wai, J. C. Yew et al., fMRI Mapping of cortical centers following visual stimulation in postnatal pigs of different ages, Life Sci, vol.78, issue.11, pp.1197-1201, 2006.

E. R. Kandel, J. H. Schwartz, and T. M. , Principles of neural science, 2012.

P. Simon, Canine Model of Convection-Enhanced Delivery of Cetuximab-Conjugated Iron-Oxide Nanoparticles Monitored With Magnetic Resonance Imaging, Neurosurgery, vol.59, issue.CN_suppl_1, pp.107-113, 2012.

E. A. Neuwelt, Osmotic blood-brain barrier disruption: pharmacodynamic studies in dogs and a clinical phase I trial in patients with malignant brain tumors, Cancer Treat. Rep, vol.65, issue.2, pp.39-43, 1981.

W. Gunnar, O. Jonasson, G. Merlotti, J. Stone, and J. Barrett, Head injury and hemorrhagic shock: studies of the blood brain barrier and intracranial pressure after resuscitation with normal saline solution, 3% saline solution, and dextran-40, Surgery, vol.103, issue.4, pp.398-407, 1988.

M. J. Russell, R. White, E. Patel, W. R. Markesbery, C. R. Watson et al., Familial influence on plaque formation in the beagle brain, Neuroreport, vol.3, issue.12, pp.1093-1096, 1992.

J. E. Rofina, Cognitive disturbances in old dogs suffering from the canine counterpart of Alzheimer's disease, Brain Res, vol.1069, issue.1, pp.216-226, 2006.

F. Schmidt, N. Willems, and A. Stolzing, Detection and Quantification of A-Amyloid, Pyroglutamyl AA, and Tau in Aged Canines, J Neuropathol Exp Neurol, vol.74, issue.9, p.12, 2015.

M. C. Osella, Canine cognitive dysfunction syndrome: prevalence, clinical signs and treatment with a neuroprotective nutraceutical, Appl. Anim. Behav. Sci, vol.105, issue.4, pp.297-310, 2007.

G. Azkona, S. Garc\'\ia-belenguer, G. Chacón, B. Rosado, M. León et al., Prevalence and risk factors of behavioural changes associated with age-related cognitive impairment in geriatric dogs, J. Small Anim. Pract, vol.50, issue.2, pp.87-91, 2009.

J. C. Neilson, B. L. Hart, K. D. Cliff, and W. W. , Prevalence of behavioral changes associated with age-related cognitive impairment in dogs, J. Am. Vet. Med. Assoc, vol.218, issue.11, pp.1787-1791, 2001.

H. E. Salvin, P. D. Mcgreevy, P. S. Sachdev, and M. J. Valenzuela, Under diagnosis of canine cognitive dysfunction: a cross-sectional survey of older companion dogs, Vet. J, vol.184, issue.3, pp.277-281, 2010.

S. Snigdha, L. Christie, C. Rivera, J. A. Araujo, N. W. Milgram et al., Age and distraction are determinants of performance on a novel visual search task in aged Beagle dogs, Age (Omaha), vol.34, issue.1, pp.67-73, 2012.

E. M. Johnstone, M. O. Chaney, F. H. Norris, R. Pascual, and S. P. Little, Conservation of the sequence of the Alzheimer's disease amyloid peptide in dog, polar bear and five other mammals by cross-species polymerase chain reaction analysis, Mol. brain Res, vol.10, issue.4, pp.299-305, 1991.

L. Sarasa, Cloning, sequencing and expression in the dog of the main amyloid precursor protein isoforms and some of the enzymes related with their processing, Neuroscience, vol.171, issue.4, pp.1091-1101, 2010.

V. Pop, A$?$ aggregation profiles and shifts in APP processing favor amyloidogenesis in canines, Neurobiol. Aging, vol.33, issue.1, pp.108-120, 2012.

J. L. Frost, Pyroglutamate-3 amyloid-$?$ deposition in the brains of humans, non-human primates, canines, and Alzheimer disease--like transgenic mouse models, Am. J. Pathol, vol.183, issue.2, pp.369-381, 2013.

B. J. Cummings, C. J. Pike, R. Shankle, and C. W. Cotman, $?$-amyloid deposition and other measures of neuropathology predict cognitive status in Alzheimer's disease, Neurobiol. Aging, vol.17, issue.6, pp.921-933, 1996.

M. Colle, Vascular and parenchymal A-? deposition in the aging dog: correlation with behavior, Neurobiol. Aging, vol.21, issue.5, pp.695-704, 2000.

T. Wisniewski, M. Lalowski, M. Bobik, M. Russell, J. Strosznajder et al., Amyloid ? 1-42 deposits do not lead to Alzheimer's neuritic plaques in aged dogs, Biochem. J, vol.313, issue.2, pp.575-580, 1996.

S. Czasch, S. Paul, and W. Baumgärtner, A comparison of immunohistochemical and silver staining methods for the detection of diffuse plaques in the aged canine brain, Neurobiol. Aging, vol.27, issue.2, pp.293-305, 2006.

M. Pugliese, J. Mascort, N. Mahy, and I. Ferrer, Diffuse beta-amyloid plaques and hyperphosphorylated tau are unrelated processes in aged dogs with behavioral deficits, Acta Neuropathol, vol.112, issue.2, pp.175-183, 2006.

K. Uchida and E. R. Stadtman, Modification of histidine residues in proteins by reaction with 4-hydroxynonenal, Proc. Natl. Acad. Sci, vol.89, pp.4544-4548, 1992.

T. Yoshino, K. Uchida, S. Tateyama, R. Yamaguchi, H. Nakayama et al., A retrospective study of canine senile plaques and cerebral amyloid angiopathy, Vet. Pathol, vol.33, issue.2, pp.230-234, 1996.

E. Head, Amyloid-? peptide and oligomers in the brain and cerebrospinal fluid of aged canines, J. Alzheimer's Dis, vol.20, issue.2, pp.637-646, 2010.

B. J. Cummings, E. Head, W. Ruehl, N. W. Milgram, and C. W. Cotman, The canine as an animal model of human aging and dementia, Neurobiol. Aging, vol.17, issue.2, pp.259-268, 1996.

M. Su, Magnetic resonance imaging of anatomic and vascular characteristics in a canine model of human aging, Neurobiol. Aging, vol.19, issue.5, pp.479-485, 1998.

P. D. Tapp, Frontal lobe volume, function, and $?$-amyloid pathology in a canine model of aging, J. Neurosci, vol.24, issue.38, pp.8205-8213, 2004.

M. J. West, C. H. Kawas, L. J. Martin, and J. C. Troncoso, The CA1 region of the human hippocampus is a hot spot in Alzheimer's disease, 2000.

C. T. Siwak-tapp, E. Head, B. A. Muggenburg, N. W. Milgram, and C. W. Cotman, Region specific neuron loss in the aged canine hippocampus is reduced by enrichment, Neurobiol. Aging, vol.29, issue.1, pp.39-50, 2008.

S. , P. Mihevc, and G. Majdi?, Canine Cognitive Dysfunction and Alzheimer's Disease -Two Facets of the Same Disease?, Front. Neurosci, vol.13, p.604, 2019.

P. C. May, The potent BACE1 inhibitor LY2886721 elicits robust central A$?$ pharmacodynamic responses in mice, dogs, and humans, J. Neurosci, vol.35, issue.3, pp.1199-1210, 2015.

W. W. Ruehl, T. L. Entriken, B. A. Muggenburg, D. S. Bruyette, W. C. Griffith et al., Treatment with L-deprenyl prolongs life in elderly dogs, Life Sci, vol.61, issue.11, pp.1037-1044, 1997.

E. Head, The effects of L-deprenyl on spatial short term memory in young and aged dogs, Prog. Neuro-Psychopharmacology Biol. Psychiatry, vol.20, issue.3, pp.515-530, 1996.

S. Campbell, A. Trettien, and B. Kozan, A noncomparative open-label study evaluating the effect of selegiline hydrochloride in a clinical setting, Vet Ther, vol.2, issue.1, pp.24-39, 2001.

C. M. Studzinski, J. A. Araujo, and N. W. Milgram, The canine model of human cognitive aging and dementia: pharmacological validity of the model for assessment of human cognitive-enhancing drugs, Prog. Neuro-Psychopharmacology Biol. Psychiatry, vol.29, issue.3, pp.489-498, 2005.

E. Barone, Biliverdin reductase-A: a novel drug target for atorvastatin in a dog pre-clinical model of Alzheimer disease, J. Neurochem, vol.120, issue.1, pp.135-146, 2012.

C. T. Siwak, Chronic antioxidant and mitochondrial cofactor administration improves discrimination learning in aged but not young dogs, Prog. Neuro-Psychopharmacology Biol. Psychiatry, vol.29, issue.3, pp.461-469, 2005.

J. A. Araujo, N. H. Greig, D. K. Ingram, J. Sandin, C. De-rivera et al., Cholinesterase inhibitors improve both memory and complex learning in aged beagle dogs, J. Alzheimer's Dis, vol.26, issue.1, pp.143-155, 2011.

U. Neumann, The BACE-1 inhibitor CNP520 for prevention trials in Alzheimer's disease, EMBO Mol. Med, vol.10, issue.11, 2018.

D. P. Dowling, A. K. Croft, and C. L. Drennan, Radical use of Rossmann and TIM barrel architectures for controlling coenzyme B12 chemistry, Annu. Rev. Biophys, vol.41, pp.403-427, 2012.

W. O. Opii, Proteomic identification of brain proteins in the canine model of human aging following a long-term treatment with antioxidants and a program of behavioral enrichment: relevance to Alzheimer's disease, Neurobiol. Aging, vol.29, issue.1, pp.51-70, 2008.

E. Head, A two-year study with fibrillar $?$-amyloid (A$?$) immunization in aged canines: effects on cognitive function and brain A$?$, J. Neurosci, vol.28, issue.14, pp.3555-3566, 2008.

P. R. Davis, A? vaccination in combination with behavioral enrichment in aged beagles: effects on cognition, A?, and microhemorrhages, Neurobiol. Aging, vol.49, pp.86-99, 2017.

M. N. Bosch, M. Pugliese, C. Andrade, J. Gimeno-bayón, N. Mahy et al., Amyloid-$?$ immunotherapy reduces amyloid plaques and astroglial reaction in aged domestic dogs, Neurodegener. Dis, vol.15, issue.1, pp.24-37, 2015.

C. T. Siwak, P. D. Tapp, and N. W. Milgram, Effect of age and level of cognitive function on spontaneous and exploratory behaviors in the beagle dog, Learn. Mem, vol.8, issue.6, pp.317-325, 2001.

A. L. Dowling and E. Head, Antioxidants in the canine model of human aging, Biochim. Biophys. Acta (BBA)-Molecular Basis Dis, vol.1822, issue.5, pp.685-689, 2012.

J. A. Araujo, G. M. Landsberg, N. W. Milgram, and A. Miolo, Improvement of short-term memory performance in aged beagles by a nutraceutical supplement containing phosphatidylserine, Ginkgo biloba, vitamin E, and pyridoxine, Can. Vet. J, vol.49, issue.4, p.379, 2008.

P. C. Huszthy, In vivo models of primary brain tumors: pitfalls and perspectives, Neuro. Oncol, vol.14, issue.8, pp.979-993, 2012.

R. B. Song, C. H. Vite, C. W. Bradley, and J. R. Cross, Postmortem evaluation of 435 cases of intracranial neoplasia in dogs and relationship of neoplasm with breed, age, and body weight, J. Vet. Intern. Med, vol.27, issue.5, pp.1143-1152, 2013.

E. P. Spugnini, D. E. Thrall, G. S. Price, N. J. Sharp, K. Munana et al., Primary irradiation of canine intracranial masses, Vet. Radiol. Ultrasound, vol.41, issue.4, pp.377-380, 2000.

J. H. , Frame-based stereotactic biopsy of canine brain masses: technique and clinical results in 26 cases, Front. Vet. Sci, vol.2, p.20, 2015.

J. H. Rossmeisl, Invited review-neuroimaging response assessment criteria for brain tumors in veterinary patients, Vet. Radiol. Ultrasound, vol.55, issue.2, pp.115-132, 2014.

Z. J. Reitman, IDH1 and IDH2 hotspot mutations are not found in canine glioma, Int. J. cancer, vol.127, issue.1, pp.245-246, 2010.

R. Thomas, Putting our heads together': insights into genomic conservation between human and canine intracranial tumors, J. Neurooncol, vol.94, issue.3, p.333, 2009.

R. J. Higgins, Spontaneous canine gliomas: overexpression of EGFR, PDGFR$?$ and IGFBP2 demonstrated by tissue microarray immunophenotyping, J. Neurooncol, vol.98, issue.1, pp.49-55, 2010.

D. York, TP53 mutations in canine brain tumors, Vet. Pathol, vol.49, issue.5, pp.796-801, 2012.

G. Stoica, H. Kim, D. G. Hall, and J. R. Coates, Morphology, immunohistochemistry, and genetic alterations in dog astrocytomas, Vet. Pathol, vol.41, issue.1, pp.10-19, 2004.

P. J. Dickinson, Canine spontaneous glioma: A translational model system for convection-enhanced delivery, Neuro. Oncol, vol.12, issue.9, pp.928-940, 2010.

T. Reya, S. J. Morrison, M. F. Clarke, and I. L. Weissman, Stem cells, cancer, and cancer stem cells, Nature, vol.414, issue.6859, p.105, 2001.

J. Rebetz, Glial progenitor-like phenotype in low-grade glioma and enhanced CD133-expression and neuronal lineage differentiation potential in high-grade glioma, PLoS One, vol.3, issue.4, p.1936, 2008.

F. Fernández, Presence of neural progenitors in spontaneous canine gliomas: A histopathological and immunohistochemical study of 20 cases, Vet. J, vol.209, pp.125-132, 2016.

M. Wrensch, Familial and personal medical history of cancer and nervous system conditions among adults with glioma and controls, Am. J. Epidemiol, vol.145, issue.7, pp.581-593, 1997.

D. Lipsitz, Glioblastoma multiforme: clinical findings, magnetic resonance imaging, and pathology in five dogs, Vet. Pathol, vol.40, issue.6, pp.659-669, 2003.

V. G. Itagiba, R. Borges, L. C. Da-cruz, A. D. Furtado, R. C. Domingues et al., Use of diffusion tensor magnetic resonance imaging in the assessment of patterns of white matter involvement in patients with brain tumors: is it useful in the differential diagnosis?, Radiol. Bras, vol.43, issue.6, pp.362-368, 2010.

R. T. Bentley, Canine intracranial gliomas: relationship between magnetic resonance imaging criteria and tumor type and grade, Vet. J, vol.198, issue.2, pp.463-471, 2013.

A. Koestner, Histological classification of tumors of the nervous system of domestic animals, World Heal. Organ. Int. Histol. Classif. Tumors Domest. Anim, pp.25-26, 1999.

A. K. Leblanc, Creation of an NCI comparative brain tumor consortium: informing the translation of new knowledge from canine to human brain tumor patients, Neuro. Oncol, vol.18, issue.9, pp.1209-1218, 2016.

J. Hicks, Intratumoral temozolomide in spontaneous canine gliomas: feasibility of a novel therapy using implanted microcylinders, Vet. Med. Sci, vol.5, issue.1, pp.5-18, 2019.

G. E. Pluhar, Anti-tumor immune response correlates with neurological symptoms in a dog with spontaneous astrocytoma treated by gene and vaccine therapy, Vaccine, vol.28, issue.19, pp.3371-3378, 2010.

A. C. Freeman, Convection-enhanced delivery of cetuximab conjugated iron-oxide nanoparticles for treatment of spontaneous canine intracranial gliomas, J. Neurooncol, vol.137, issue.3, pp.653-663, 2018.

W. Xiong, Human Flt3L generates dendritic cells from canine peripheral blood precursors: implications for a dog glioma clinical trial, PLoS One, vol.5, issue.6, p.11074, 2010.

K. M. Kendrick, A. P. Da-costa, A. E. Leigh, M. R. Hinton, and J. W. Peirce, Sheep don't forget a face, Nature, vol.414, issue.6860, p.165, 2001.

G. T. Truong, Role of nitrite in regulation of fetal cephalic circulation in sheep, J. Physiol, vol.592, issue.8, pp.1785-1794, 2014.

H. I. Opdam, A sheep model for the study of focal epilepsy with concurrent intracranial EEG and functional MRI, Epilepsia, vol.43, issue.8, pp.779-787, 2002.

J. Boltze, Permanent middle cerebral artery occlusion in sheep: a novel large animal model of focal cerebral ischemia, J. Cereb. Blood Flow Metab, vol.28, issue.12, pp.1951-1964, 2008.

A. J. Wells, A surgical model of permanent and transient middle cerebral artery stroke in the sheep, PLoS One, vol.7, issue.7, p.42157, 2012.

N. A. Terpolilli, Inhalation of nitric oxide prevents ischemic brain damage in experimental stroke by selective dilatation of collateral arterioles, Circ. Res, vol.110, issue.5, pp.727-738, 2012.

A. Grimmelt, Closed traumatic brain injury model in sheep mimicking high-velocity, closed head trauma in humans, Cent. Eur. Neurosurgery-Zentralblatt für Neurochir, vol.72, issue.03, pp.120-126, 2011.

R. D. Jolly, D. G. Arthur, G. W. Kay, and D. N. Palmer, Neuronal ceroid-lipofuscinosis in Borderdale sheep, N. Z. Vet. J, vol.50, issue.5, pp.199-202, 2002.

M. Bond, S. Holthaus, I. Tammen, G. Tear, and C. Russell, Use of model organisms for the study of neuronal ceroid lipofuscinosis, Biochim. Biophys. Acta (BBA)-Molecular Basis Dis, vol.1832, issue.11, pp.1842-1865, 2013.

N. Hunter, Transmission of prion diseases by blood transfusion, J. Gen. Virol, vol.83, issue.11, pp.2897-2905, 2002.

J. C. Jacobsen, An ovine transgenic Huntington's disease model, Hum. Mol. Genet, vol.19, issue.10, pp.1873-1882, 2010.

N. Perentos, Translational neurophysiology in sheep: measuring sleep and neurological dysfunction in CLN5 Batten disease affected sheep, Brain, vol.138, issue.4, pp.862-874, 2015.

R. D. Geraets, S. Koh, M. L. Hastings, T. Kielian, D. A. Pearce et al., Moving towards effective therapeutic strategies for Neuronal Ceroid Lipofuscinosis, Orphanet J. Rare Dis, vol.11, issue.1, p.40, 2016.

A. R. Pinnapureddy, C. Stayner, J. Mcewan, O. Baddeley, J. Forman et al., Large animal models of rare genetic disorders: sheep as phenotypically relevant models of human genetic disease, Orphanet J. Rare Dis, vol.10, issue.1, p.107, 2015.

D. N. Palmer, Recent studies of ovine neuronal ceroid lipofuscinoses from BARN, the Batten Animal Research Network, Biochim. Biophys. Acta (BBA)-Molecular Basis Dis, vol.1852, issue.10, pp.2279-2286, 2015.

S. B. Prusiner, Prions, Proc. Natl. Acad. Sci, vol.95, issue.23, pp.13363-13383, 1998.

J. S. Griffith, Nature of the scrapie agent: Self-replication and scrapie, Nature, vol.215, issue.5105, p.1043, 1967.

S. B. Prusiner, Novel proteinaceous infectious particles cause scrapie, Science (80-. ), vol.216, issue.4542, pp.136-144, 1982.

M. E. Bruce, Strain characterization of natural sheep scrapie and comparison with BSE, J. Gen. Virol, vol.83, issue.3, pp.695-704, 2002.

P. Parchi, Classification of sporadic Creutzfeldt-Jakob disease based on molecular and phenotypic analysis of 300 subjects, Ann. Neurol, vol.46, issue.2, pp.224-233, 1999.

S. L. Benestad, J. Arsac, W. Goldmann, and M. Nöremark, Atypical/Nor98 scrapie: properties of the agent, genetics, and epidemiology, Vet. Res, vol.39, issue.4, pp.1-14, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00390572

S. L. Benestad, P. Sarradin, B. Thu, J. Schönheit, M. A. Tranulis et al., Cases of scrapie with unusual features in Norway and designation of a new type, Nor98, Vet. Rec, vol.153, issue.7, pp.202-208, 2003.

J. Arsac, O. Andreoletti, J. Bilheude, C. Lacroux, S. L. Benestad et al., Similar biochemical signatures and prion protein genotypes in atypical scrapie and Nor98 cases, France and Norway, Emerg. Infect. Dis, vol.13, issue.1, p.58, 2007.

P. Vitali, Diffusion-weighted MRI hyperintensity patterns differentiate CJD from other rapid dementias, Neurology, vol.76, issue.20, pp.1711-1719, 2011.

E. Caverzasi, Application of quantitative DTI metrics in sporadic CJD, NeuroImage Clin, vol.4, pp.426-435, 2014.

G. S. Young, Diffusion-weighted and fluid-attenuated inversion recovery imaging in Creutzfeldt-Jakob disease: high sensitivity and specificity for diagnosis, Am. J. Neuroradiol, vol.26, issue.6, pp.1551-1562, 2005.

M. D. Geschwind, Correlating DWI MRI with pathological and other features of Jakob-Creutzfeldt disease, Alzheimer Dis. Assoc. Disord, vol.23, issue.1, p.82, 2009.

R. B. Mars, Primate comparative neuroscience using magnetic resonance imaging: promises and challenges, Front. Neurosci, vol.8, p.298, 2014.

H. Johansen-berg and M. F. Rushworth, Using diffusion imaging to study human connectional anatomy, Annu. Rev. Neurosci, vol.32, pp.75-94, 2009.

S. Jbabdi and H. Johansen-berg, Tractography: where do we go from here?, Brain Connect, vol.1, issue.3, pp.169-183, 2011.

R. B. Mars, Diffusion-weighted imaging tractography-based parcellation of the human parietal cortex and comparison with human and macaque resting-state functional connectivity, J. Neurosci, vol.31, issue.11, pp.4087-4100, 2011.

M. S. Anaya-garc\'\ia, J. S. Anaya, O. Meléndez, J. L. Velázquez-ram\'\irez, and R. Aguiar, In Vivo study of cerebral white matter in the dog using diffusion tensor tractography, Vet. Radiol. Ultrasound, vol.56, issue.2, pp.188-195, 2015.

H. L. Gray-edwards, High resolution MRI anatomy of the cat brain at 3 Tesla, J. Neurosci. Methods, vol.227, pp.10-17, 2014.

K. Stuckenschneider, M. Hellige, K. Feige, and H. Gasse, 3-Tesla magnetic resonance imaging of the equine brain in healthy horses -Potentials and limitations, Pferdeheilkd. Equine Med, vol.30, issue.6, pp.657-670, 2014.

M. J. Schmidt, C. Knemeyer, and H. Heinsen, Neuroanatomy of the equine brain as revealed by highfield (3Tesla) magnetic-resonance-imaging, PLoS One, vol.14, issue.4, p.213814, 2019.

D. Kim, In vivo mapping of functional domains and axonal connectivity in cat visual cortex using magnetic resonance imaging, Magn. Reson. Imaging, vol.21, issue.10, pp.1131-1140, 2003.

J. Zhong, An in vivo multi-modal structural template for neonatal piglets using high angular resolution and population-based whole-brain tractography, Front. Neuroanat, vol.10, p.92, 2016.

W. Lee, Functional and diffusion tensor magnetic resonance imaging of the sheep brain, BMC Vet. Res, vol.11, issue.1, 2015.

O. Jacqmot, Diffusion tensor imaging of white matter tracts in the dog brain, Anat. Rec, vol.296, issue.2, pp.340-349, 2013.

J. L. Robinson, Characterization of Structural Connectivity of the Default Mode Network in Dogs using Diffusion Tensor Imaging, Sci. Rep, vol.6, issue.1, 2016.

S. P. Kyathanahally, Anterior--posterior dissociation of the default mode network in dogs, Brain Struct. Funct, vol.220, issue.2, pp.1063-1076, 2015.

G. E. Alexander, Age-related regional network of magnetic resonance imaging gray matter in the rhesus macaque, J. Neurosci, vol.28, issue.11, pp.2710-2718, 2008.

T. R. Oakes, A. S. Fox, T. Johnstone, M. K. Chung, N. Kalin et al., Integrating VBM into the General Linear Model with voxelwise anatomical covariates, Neuroimage, vol.34, issue.2, pp.500-508, 2007.

D. G. Mclaren, K. J. Kosmatka, E. K. Kastman, B. B. Bendlin, and S. C. Johnson, Rhesus macaque brain morphometry: a methodological comparison of voxel-wise approaches, Methods, vol.50, issue.3, pp.157-165, 2010.

B. Nie, H. Liu, K. Chen, X. Jiang, and B. Shan, A statistical parametric mapping toolbox used for voxelwise analysis of FDG-PET images of rat brain, PLoS One, vol.9, issue.9, p.108295, 2014.

J. Lebenberg, A. Hérard, A. Dubois, M. Dhenain, P. Hantraye et al., A combination of atlas-based and voxel-wise approaches to analyze metabolic changes in autoradiographic data from Alzheimer's mice, Neuroimage, vol.57, issue.4, pp.1447-1457, 2011.

A. Ella and M. Keller, Construction of an MRI 3D high resolution sheep brain template, Magn. Reson. Imaging, vol.33, issue.10, pp.1329-1337, 2015.

A. Ella, J. A. Delgadillo, P. Chemineau, and M. Keller, Computation of a high-resolution MRI 3D stereotaxic atlas of the sheep brain, J. Comp. Neurol, vol.525, issue.3, pp.676-692, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01433685

P. D. Tapp, K. Head, E. Head, N. W. Milgram, B. A. Muggenburg et al., Application of an automated voxel-based morphometry technique to assess regional gray and white matter brain atrophy in a canine model of aging, Neuroimage, 2006.

R. Datta, A digital atlas of the dog brain, PLoS One, vol.7, issue.12, p.52140, 2012.

M. E. Milne, C. Steward, S. M. Firestone, S. N. Long, T. J. O'brien et al., Development of representative magnetic resonance imaging--based atlases of the canine brain and evaluation of three methods for atlas-based segmentation, Am. J. Vet. Res, vol.77, issue.4, pp.395-403, 2016.

B. B. Avants, N. Tustison, and G. Song, Advanced Normalization Tools (ANTS), Insight J, pp.1-35, 2009.

B. B. Avants, The optimal template effect in hippocampus studies of diseased populations, Neuroimage, vol.49, issue.3, pp.2457-2466, 2010.

J. Yelnik, A three-dimensional, histological and deformable atlas of the human basal ganglia. I. Atlas construction based on immunohistochemical and MRI data, Neuroimage, 2007.
URL : https://hal.archives-ouvertes.fr/inria-00616029

P. Bazin and D. L. Pham, Homeomorphic brain image segmentation with topological and statistical atlases, Med. Image Anal, vol.12, issue.5, pp.616-625, 2008.

R. Datta, A Digital Atlas of the Dog Brain, PLoS One, 2012.

L. Risser, A. Sadoun, M. Mescam, K. Strelnikov, S. Lebreton et al., In vivo localization of cortical areas using a 3D computerized atlas of the marmoset brain, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02327022

X. Palazzi, The beagle brain in stereotaxic coordinates, The Beagle Brain in Stereotaxic Coordinates, pp.1-115, 2011.

K. J. Black, A. Z. Snyder, J. M. Koller, M. H. Gado, and J. S. Perlmutter, Template Images for Nonhuman Primate Neuroimaging: 1. Baboon, 2001.

A. Ella and M. Keller, Construction of an MRI 3D high resolution sheep brain template, Magn. Reson. Imaging, vol.33, issue.10, pp.1329-1337, 2015.

T. Hashikawa, R. Nakatomi, and A. Iriki, Current models of the marmoset brain, Neurosci. Res, vol.93, pp.116-127, 2015.

D. G. Mclaren, A population-average MRI-based atlas collection of the rhesus macaque, Neuroimage, 2009.

D. Lingrand, , 2008.

Y. Assaf and O. Pasternak, Diffusion Tensor Imaging (DTI)-based White Matter Mapping in Brain Research: A Review, 2007.

M. Catani and M. Thiebaut-de-schotten, A diffusion tensor imaging tractography atlas for virtual in vivo dissections, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00322865

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

M. Daianu, An advanced white matter tract analysis in frontotemporal dementia and early-onset Alzheimer's disease, Brain Imaging Behav, vol.10, pp.1038-1053, 2016.

V. Agarwal, J. G. Malcolm, G. Pradilla, and D. L. Barrow, Tractography for Optic Radiation Preservation in Transcortical Approaches to Intracerebral Lesions, Cureus, 2017.

W. Lee, B. Park, and K. Han, Classification of diffusion tensor images for the early detection of Alzheimer's disease, Comput. Biol. Med, vol.43, pp.1313-1320, 2013.

W. Tan, Deterministic Tractography of the Nigrostriatal-Nigropallidal Pathway in Parkinson's Disease, Sci. Rep, vol.5, issue.1, 2015.

J. Berberat, J. Mcnamara, L. Remonda, S. Bodis, and S. Rogers, Diffusion tensor imaging for target volume definition in glioblastoma multiforme, Strahlenther Onkol, vol.190, pp.939-943, 2014.

M. Urbanski, DTI-MR tractography of white matter damage in stroke patients with neglect, Exp Brain Res, vol.208, pp.491-505, 2011.

M. Moller, J. Frandsen, G. Andersen, A. Gjedde, P. Vestergaard-poulsen et al., Dynamic changes in corticospinal tracts after stroke detected by fibretracking, J. Neurol. Neurosurg. Psychiatry, vol.78, issue.6, pp.587-592, 2007.

S. Cho, D. G. Kim, D. Kim, Y. Kim, C. Lee et al., Motor outcome according to the integrity of the corticospinal tract determined by diffusion tensor tractography in the early stage of corona radiata infarct, Neurosci. Lett, vol.426, pp.123-127, 2007.

S. Lee, J. Coutu, P. Wilkens, A. Yendiki, H. D. Rosas et al., Tract-based analysis of white matter degeneration in Alzheimer's disease, Neuroscience, vol.301, pp.79-89, 2015.

M. S. Anaya-garcía, J. S. Anaya, O. Meléndez, J. L. Velázquez-ramírez, and R. Aguiar, VIVO STUDY OF CEREBRAL WHITE MATTER IN THE DOG USING DIFFUSION TENSOR TRACTOGRAPHY, vol.56, pp.188-195, 2015.

H. L. Gray-edwards, High resolution MRI anatomy of the cat brain at 3 Tesla, J. Neurosci. Methods, vol.227, issue.227, pp.10-17, 2014.

K. Stuckenschneider, M. Hellige, K. Feige, and H. Gasse, 3-Tesla magnetic resonance imaging of the equine brain in healthy horses -Potentials and limitations, Pferdeheilkd. Equine Med, vol.30, issue.6, pp.657-670, 2014.

W. Lee, Functional and diffusion tensor magnetic resonance imaging of the sheep brain, BMC Vet. Res, issue.11, 2012.

O. Jacqmot, Diffusion Tensor Imaging of White Matter Tracts in the Dog Brain

J. L. Robinson, Characterization of Structural Connectivity of the Default Mode Network in Dogs using Diffusion Tensor Imaging, 2016.

A. Das and E. Takahashi, Characterization of White Matter Tracts by Diffusion MR Tractography in Cat and Ferret that Have Similar Gyral Patterns, Cereb. Cortex, vol.28, issue.4, pp.1338-1347, 2018.

E. Takahashi, Development of cerebral fiber pathways in cats revealed by diffusion spectrum imaging, Neuroimage, vol.49, issue.2, pp.1231-1240, 2010.

J. K. Chambers, The domestic cat as a natural animal model of Alzheimer's disease, Acta Neuropathol. Commun, vol.3, issue.1, 2015.

D. Mcfarlane, Advantages and limitations of the equine disease, pituitary pars intermedia dysfunction as a model of spontaneous dopaminergic neurodegenerative disease, Ageing Res. Rev, vol.6, issue.1, pp.54-63, 2007.

H. T. Chang, W. K. Rumbeiha, J. S. Patterson, B. Puschner, and A. P. Knight, Toxic Equine Parkinsonism: An Immunohistochemical Study of 10 Horses With Nigropallidal Encephalomalacia, Vet. Pathol, vol.49, issue.2, pp.398-402, 2012.

A. M. Bradbury, B. L. Gurda, M. L. Casal, K. P. Ponder, C. H. Vite et al., A Review of Gene Therapy in Canine and Feline Models of Lysosomal Storage Disorders

L. Karageorgos, M. J. Lancaster, J. S. Nimmo, and J. J. Hopwood, Gaucher disease in sheep, J. Inherit. Metab. Dis, vol.34, issue.1, pp.209-215, 2011.

R. Pascalau, C. C. Aldea, V. A. Padurean, and B. Szabo, Comparative Study of the Major White Matter Tracts Anatomy in Equine, Feline and Canine Brains by Use of the Fibre Dissection Technique, J. Vet. Med. Ser. C Anat. Histol. Embryol, vol.45, issue.5, pp.373-385, 2016.

A. S. Shatil, K. M. Matsuda, and C. R. Figley, A method for whole brain ex vivo magnetic resonance imaging with minimal susceptibility artifacts, Front. Neurol, vol.7, issue.NOV, pp.1-10, 2016.

B. B. Avants, N. J. Tustison, G. Song, P. A. Cook, A. Klein et al., A reproducible evaluation of ANTs similarity metric performance in brain image registration, Neuroimage, vol.54, pp.2033-2044, 2011.

P. J. Basser, J. Mattiello, and D. Lebihan, MR diffusion tensor spectroscopy and imaging, Biophys. J, vol.66, issue.1, pp.259-67, 1994.
URL : https://hal.archives-ouvertes.fr/hal-00349721

F. Yeh, T. D. Verstynen, Y. Wang, J. C. Fernández-miranda, and W. Tseng, Deterministic Diffusion Fiber Tracking Improved by Quantitative Anisotropy, PLoS One, vol.8, issue.11, 2013.

M. S. Anaya-garca, VIVO STUDY OF CEREBRAL WHITE MATTER IN THE DOG USING DIFFUSION TENSOR TRACTOGRAPHY

Y. Bao, Y. Wang, W. Wang, and Y. Wang, The Superior Fronto-Occipital Fasciculus in the Human Brain Revealed by Diffusion Spectrum Imaging Tractography: An Anatomical Reality or a Methodological Artifact?, Front. Neuroanat, vol.11, pp.1-10, 2018.

A. Meola, A. Comert, F. C. Yeh, L. Stefaneanu, and J. C. Fernandez-miranda, The controversial existence of the human superior fronto-occipital fasciculus: Connectome-based tractographic study with microdissection validation, Hum. Brain Mapp, vol.36, issue.12, pp.4964-4971, 2015.

E. Caverzasi, White matter involvement in sporadic Creutzfeldt-Jakob disease, Brain, vol.137, issue.12, pp.3339-3354, 2014.

J. Manjón, P. Coupé, L. Concha, A. Buades, D. L. Collins et al., Diffusion Weighted Image Denoising Using Overcomplete Local PCA, 2013.

A. Moussavi-biugui, B. Stieltjes, K. Fritzsche, W. Semmler, and F. B. Laun, Novel spherical phantoms for Q-ball imaging under in vivo conditions, Magn. Reson. Med, vol.65, issue.1, pp.190-194, 2011.

M. Descoteaux, E. Angelino, S. Fitzgibbons, and R. Deriche, Regularized, fast, and robust analytical Qball imaging, Magn. Reson. Med, vol.58, issue.3, pp.497-510, 2007.