R. Gardner, J. Burke, J. Nettiksimmons, A. Kaup, and D. Barnes, Dementia Risk After Traumatic Brain Injury vs Nonbrain Trauma, JAMA Neurology, vol.71, issue.12, pp.1490-1497, 2014.
DOI : 10.1001/jamaneurol.2014.2668

D. Katz, S. Cohen, and M. Alexander, Mild traumatic brain injury, Handb Clin Neuro, vol.127, pp.131-156, 2015.
DOI : 10.1016/B978-0-444-52892-6.00009-X

K. Marshall, S. Holland, K. Meyer, E. Martin, and M. Wilmore, Mild Traumatic Brain Injury Screening, Diagnosis, and Treatment, Military Medicine, vol.177, issue.8S, pp.67-75, 2012.
DOI : 10.7205/MILMED-D-12-00110

J. Niemeier, L. Grafton, and T. Chilakamarri, Treating Persons With Traumatic Brain Injury, North Carolina Medical Journal, vol.76, issue.2, pp.105-110, 2015.
DOI : 10.18043/ncm.76.2.105

N. Chauhan, Chronic neurodegenerative consequences of traumatic brain injury, Restor Neurol Neurosci, vol.32, pp.337-365, 2014.

B. Masel and D. Dewitt, Traumatic Brain Injury: A Disease Process, Not an Event, Journal of Neurotrauma, vol.27, issue.8, pp.1529-1540, 2010.
DOI : 10.1089/neu.2010.1358

V. Johnson, W. Stewart, and D. Smith, Widespread Tau and Amyloid-Beta Pathology Many Years After a Single Traumatic Brain Injury in Humans, Brain Pathology, vol.208, issue.2, pp.142-149, 2012.
DOI : 10.1111/j.1750-3639.2011.00513.x

K. Uryu, H. Laurer, T. Mcintosh, D. Praticò, and D. Martinez, Repetitive mild brain trauma accelerates Abeta deposition, lipid peroxidation, and cognitive impairment in a transgenic mouse model of Alzheimer amyloidosis, J Neurosci, vol.22, pp.446-454, 2002.

M. Lamprecht, B. Elkin, K. Kesavabhotla, J. Crary, and J. Hammers, Implicates a Role for SORLA, Journal of Neurotrauma, vol.34, issue.1, 2016.
DOI : 10.1089/neu.2015.4306

J. Burke, J. Stulc, L. Skolarus, E. Sears, and D. Zahuranec, Traumatic brain injury may be an independent risk factor for stroke, Neurology, vol.81, issue.1, pp.33-39, 2013.
DOI : 10.1212/WNL.0b013e318297eecf

N. Osier and C. Dixon, Catecholaminergic based therapies for functional recovery after TBI, Brain Research, vol.1640, pp.15-35, 2015.
DOI : 10.1016/j.brainres.2015.12.026

E. Carré, E. Cantais, O. Darbin, J. Terrier, and M. Lonjon, Technical aspects of an impact acceleration traumatic brain injury rat model with potential suitability for both microdialysis and PtiO2 monitoring, Journal of Neuroscience Methods, vol.140, issue.1-2, pp.23-28, 2004.
DOI : 10.1016/j.jneumeth.2004.04.037

S. Kabadi, G. Hilton, B. Stoica, D. Zapple, and A. Faden, Fluid-percussion???induced traumatic brain injury model in rats, Nature Protocols, vol.1, issue.9, pp.1552-1563, 2010.
DOI : 10.1038/nprot.2010.112

Y. Xiong, A. Mahmood, and M. Chopp, Animal models of traumatic brain injury, Nature Reviews Neuroscience, vol.2, issue.2, pp.128-142, 2013.
DOI : 10.1038/nrn3407

M. Risling and J. Davidsson, Experimental Animal Models for Studies on the Mechanisms of Blast-Induced Neurotrauma, Frontiers in Neurology, vol.3, p.30, 2012.
DOI : 10.3389/fneur.2012.00030

T. Colak, N. Cine, B. Bamac, O. Kurtas, and A. Ozbek, Microarray-based gene expression analysis of an animal model for closed head injury, Injury, vol.43, issue.8, pp.1264-1270, 2012.
DOI : 10.1016/j.injury.2012.01.021

M. Risling, S. Plantman, M. Angeria, E. Rostami, and B. Bellander, Mechanisms of blast induced brain injuries, experimental studies in rats, NeuroImage, vol.54, issue.1, pp.89-97, 2011.
DOI : 10.1016/j.neuroimage.2010.05.031

M. Sabir, P. Gaudreault, M. Freyburger, R. Massart, and A. Blanchet-cohen, Impact of traumatic brain injury on sleep structure, electrocorticographic activity and transcriptome in mice, Brain, Behavior, and Immunity, vol.47, pp.118-130, 2015.
DOI : 10.1016/j.bbi.2014.12.023

H. Shojo, Y. Kaneko, T. Mabuchi, K. Kibayashi, and N. Adachi, Genetic and histologic evidence implicates role of inflammation in traumatic brain injury-induced apoptosis in the rat cerebral cortex following moderate fluid percussion injury, Neuroscience, vol.171, issue.4, pp.1273-1282, 2010.
DOI : 10.1016/j.neuroscience.2010.10.018

C. Von-gertten, F. Morales, A. Holmin, S. Mathiesen, T. Nordqvist et al., Genomic responses in rat cerebral cortex after traumatic brain injury, BMC, vol.6, p.69, 2005.

T. White, M. Surles-zeigler, G. Ford, A. Gates, and B. Davids, Bilateral gene interaction hierarchy analysis of the cell death gene response emphasizes the significance of cell cycle genes following unilateral traumatic brain injury, BMC Genomics, vol.21, issue.9, p.130, 2016.
DOI : 10.1186/s12864-016-2412-0

J. Dalley, R. Cardinal, and T. Robbins, Prefrontal executive and cognitive functions in rodents: neural and neurochemical substrates, Neuroscience & Biobehavioral Reviews, vol.28, issue.7, pp.771-784, 2004.
DOI : 10.1016/j.neubiorev.2004.09.006

T. Mcallister, M. Sparling, L. Flashman, S. Guerin, and A. Mamourian, Differential Working Memory Load Effects after Mild Traumatic Brain Injury, NeuroImage, vol.14, issue.5, pp.1004-1012, 2001.
DOI : 10.1006/nimg.2001.0899

V. Paban, Molecular gene expression following blunt and rotational models of traumatic brain injury parallel injuries associated with stroke and depression J Transl Sci, 2016 doi: 10.15761/JTS, pp.330-339, 2016.

G. Turner, A. Mcintosh, and B. Levine, Prefrontal Compensatory Engagement in TBI is due to Altered Functional Engagement Of Existing Networks and not Functional Reorganization, Frontiers in Systems Neuroscience, vol.5, pp.9-12, 2011.
DOI : 10.3389/fnsys.2011.00009

J. Davidsson and M. Risling, A New Model to Produce Sagittal Plane Rotational Induced Diffuse Axonal Injuries, Frontiers in Neurology, vol.2, p.41, 2011.
DOI : 10.3389/fneur.2011.00041

URL : http://doi.org/10.3389/fneur.2011.00041

G. Paxinos and C. Watson, The rat brain in stereotaxic coordinates, 1998.

Y. Benjamini, D. Drai, G. Elmer, N. Kafkafi, and I. Golani, Controlling the false discovery rate in behavior genetics research, Behavioural Brain Research, vol.125, issue.1-2, pp.279-284, 2001.
DOI : 10.1016/S0166-4328(01)00297-2

A. Reiner, D. Yekutieli, and Y. Benjamini, Identifying differentially expressed genes using false discovery rate controlling procedures, Bioinformatics, vol.19, issue.3, pp.368-375, 2003.
DOI : 10.1093/bioinformatics/btf877

V. Paban, C. Chambon, F. Farioli, and B. Alescio-lautier, Gene regulation in the rat prefrontal cortex after learning with or without cholinergic insult, Neurobiology of Learning and Memory, vol.95, issue.4, pp.441-452, 2011.
DOI : 10.1016/j.nlm.2011.02.005

URL : https://hal.archives-ouvertes.fr/hal-01440422

V. Paban, F. Farioli, B. Romier, C. Chambon, and B. Alescio-lautier, Gene expression profile in rat hippocampus with and without memory deficit, Neurobiology of Learning and Memory, vol.94, issue.1, pp.42-56, 2010.
DOI : 10.1016/j.nlm.2010.03.005

URL : https://hal.archives-ouvertes.fr/hal-01440467

C. Benatti, C. Valensisi, J. Blom, S. Alboni, and C. Montanari, Transcriptional profiles underlying vulnerability and resilience in rats exposed to an acute unavoidable stress, Journal of Neuroscience Research, vol.35, issue.Suppl 1, pp.2103-2115, 2012.
DOI : 10.1002/jnr.23100

E. Blaveri, F. Kelly, A. Mallei, K. Harris, and A. Taylor, Expression Profiling of a Genetic Animal Model of Depression Reveals Novel Molecular Pathways Underlying Depressive-Like Behaviours, PLoS ONE, vol.106, issue.9, p.12596, 2010.
DOI : 10.1371/journal.pone.0012596.s005

M. Kimpel, W. Strother, J. Mcclintick, L. Carr, and T. Liang, Functional gene expression differences between inbred alcohol-preferring and ???non-preferring rats in five brain regions, Alcohol, vol.41, issue.2, pp.95-132, 2007.
DOI : 10.1016/j.alcohol.2007.03.003

J. Ramos-cejudo, M. Gutiérrez-fernández, B. Rodríguez-frutos, M. Expósitoalcaide, and F. Sánchez-cabo, Spatial and Temporal Gene Expression Differences in Core and Periinfarct Areas in Experimental Stroke: A Microarray Analysis, PLoS ONE, vol.18, issue.12, p.52121, 2012.
DOI : 10.1371/journal.pone.0052121.s006

K. Wirz, K. Bossers, A. Stargardt, W. Kamphuis, and D. Swaab, Cortical beta amyloid protein triggers an immune response, but no synaptic changes in the APPswe/PS1dE9 Alzheimer's disease mouse model, Neurobiology of Aging, vol.34, issue.5, pp.1328-1342, 2013.
DOI : 10.1016/j.neurobiolaging.2012.11.008

E. Blalock, R. Grondin, K. Chen, O. Thibault, and V. Thibault, Aging-Related Gene Expression in Hippocampus Proper Compared with Dentate Gyrus Is Selectively Associated with Metabolic Syndrome Variables in Rhesus Monkeys, Journal of Neuroscience, vol.30, issue.17, pp.6058-6071, 2010.
DOI : 10.1523/JNEUROSCI.3956-09.2010

W. Huang-da, B. Sherman, and R. Lempicki, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists, Nucleic Acids Research, vol.37, issue.1, pp.1-13, 2009.
DOI : 10.1093/nar/gkn923

W. Huang-da, B. Sherman, and R. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nature Protocols, vol.99, issue.1, pp.44-57, 2009.
DOI : 10.1038/nprot.2008.211

J. Mehta and R. S. , Software and Tools for Microarray Data Analysis, Methods MolBiol, vol.784, pp.41-53, 2011.
DOI : 10.1007/978-1-61779-289-2_4

J. Montojo, K. Zuberi, H. Rodriguez, G. Bader, and Q. Morris, GeneMANIA: Fast gene network construction and function prediction for Cytoscape, F1000Research, 2014.
DOI : 10.12688/f1000research.4572.1

P. Dash, N. Kobori, and A. Moore, A Molecular Description of Brain Trauma Pathophysiology Using Microarray Technology: An Overview, Neurochemical Research, vol.29, issue.6, pp.1275-1286, 2004.
DOI : 10.1023/B:NERE.0000023614.30084.eb

Y. Long, L. Zou, H. Liu, H. Lu, and X. Yuan, Altered expression of randomly selected genes in mouse hippocampus after traumatic brain injury, Journal of Neuroscience Research, vol.71, issue.5, pp.710-720, 2003.
DOI : 10.1002/jnr.10524

J. Natale, F. Ahmed, I. Cernak, B. Stoica, and A. Faden, Gene Expression Profile Changes Are Commonly Modulated across Models and Species after Traumatic Brain Injury, Journal of Neurotrauma, vol.20, issue.10, pp.907-927, 2003.
DOI : 10.1089/089771503770195777

X. Zhang, C. Gu, J. Gu, J. Zhang, and H. Zhu, Analysis of key genes and modules during the courses of traumatic brain injury with microarray technology, Genetics and Molecular Research, vol.13, issue.4, 2014.
DOI : 10.4238/2014.November.7.9

N. Kobori, G. Clifton, and P. Dash, Altered expression of novel genes in the cerebral cortex following experimental brain injury, Molecular Brain Research, vol.104, issue.2, pp.148-158, 2002.
DOI : 10.1016/S0169-328X(02)00331-5

D. Tweedie, L. Rachmany, D. Kim, V. Rubovitch, and E. Lehrmann, Mild traumatic brain injury-induced hippocampal gene expressions: The identification of target cellular processes for drug development, Journal of Neuroscience Methods, vol.272, 2016.
DOI : 10.1016/j.jneumeth.2016.02.003

. Darkazallia, C. Vied, C. Badger, and C. Levenson, Human Mesenchymal Stem Cell Treatment Normalizes Cortical Gene Expression after Traumatic Brain Injury, Journal of Neurotrauma, vol.34, issue.1, 2016.
DOI : 10.1089/neu.2015.4322

C. Kim, M. Nakamura, and C. Hsieh, Brain trauma elicits non-canonical macrophage activation states, Journal of Neuroinflammation, vol.30, issue.20, p.117, 2016.
DOI : 10.1186/s12974-016-0581-z

K. Merchant-borna, H. Lee, D. Wang, V. Bogner, and M. Van-griensven, Genome-Wide Changes in Peripheral Gene Expression following Sports-Related Concussion, Journal of Neurotrauma, vol.33, issue.17, pp.1576-1585, 2016.
DOI : 10.1089/neu.2015.4191

N. Lon?arevi?-vasiljkovi?, D. Milanovi?, V. Pe?i?, V. Te?i?, and M. Brki?, Dietary restriction suppresses apoptotic cell death, promotes Bcl-2 and Bcl-xl mRNA expression and increases the Bcl-2/Bax protein ratio in the rat cortex after cortical injury, Neurochemistry International, vol.96, pp.69-76, 2016.
DOI : 10.1016/j.neuint.2016.02.017

P. Dash, G. Hergenroeder, C. Jeter, H. Choi, and N. Kobori, Traumatic Brain Injury Alters Methionine Metabolism: Implications for Pathophysiology, Frontiers in Systems Neuroscience, vol.429, p.36, 2016.
DOI : 10.1016/j.neulet.2007.09.061

URL : http://doi.org/10.3389/fnsys.2016.00036

Y. Cho, L. Latour, H. Kim, L. Turtzo, and A. Olivera, Older Age Results in Differential Gene Expression after Mild Traumatic Brain Injury and Is Linked to Imaging Differences at Acute Follow-up, Frontiers in Aging Neuroscience, vol.51, issue.18, p.168, 2016.
DOI : 10.1016/j.nurt.2009.10.016

M. Heinzelmann, S. Reddy, L. French, D. Wang, and H. Lee, Military Personnel with Chronic Symptoms Following Blast Traumatic Brain Injury Have Differential Expression of Neuronal Recovery and Epidermal Growth Factor Receptor Genes, Frontiers in Neurology, vol.12, issue.8, p.98, 2014.
DOI : 10.1016/j.dnarep.2013.04.011

B. Stemper, A. Shah, F. Pintar, M. Mccrea, and S. Kurpad, Head Rotational Acceleration Characteristics Influence Behavioral and Diffusion Tensor Imaging Outcomes Following Concussion, Annals of Biomedical Engineering, vol.10, issue.5, pp.1071-1088, 2015.
DOI : 10.1007/s10439-014-1171-9

R. Jorge, Mood disorders, Handb Clin Neurol, vol.128, pp.613-631, 2015.
DOI : 10.1016/B978-0-444-63521-1.00038-8

D. Zgaljardic, G. Seale, L. Schaefer, R. Temple, and J. Foreman, Psychiatric Disease and Post-Acute Traumatic Brain Injury, Journal of Neurotrauma, vol.32, issue.23, pp.1911-1925, 2015.
DOI : 10.1089/neu.2014.3569

W. Marsden, Synaptic plasticity in depression: Molecular, cellular and functional correlates, Progress in Neuro-Psychopharmacology and Biological Psychiatry, vol.43, pp.168-184, 2013.
DOI : 10.1016/j.pnpbp.2012.12.012

D. Martins-de-souza, P. Guest, L. Harris, N. Vanattou-saifoudine, and M. Webster, Identification of proteomic signatures associated with depression and psychotic depression in post-mortem brains from major depression patients, Translational Psychiatry, vol.69, issue.3, p.87, 2012.
DOI : 10.1038/tp.2012.13

L. Morries, P. Cassano, and T. Henderson, Treatments for traumatic brain injury with emphasis on transcranial near-infrared laser phototherapy, Neuropsychiatr Dis Treat, vol.11, pp.2159-2175, 2015.

L. Xie, J. Calafat, H. Janssen, J. De-la-iglesia-vicente, and F. Mollinedo, Intracellular location of syntaxin 7 in human neutrophils, Immunology Letters, vol.129, issue.2, pp.72-77, 2010.
DOI : 10.1016/j.imlet.2010.02.003

Y. Pan and L. Xia, Emerging roles of podoplanin in vascular development and homeostasis, Frontiers of Medicine, vol.31, issue.9, pp.421-430, 2015.
DOI : 10.1007/s11684-015-0424-9

E. Kokovay, Y. Wang, G. Kusek, R. Wurster, and P. Lederman, VCAM1 Is Essential to Maintain the Structure of the SVZ Niche and Acts as an Environmental Sensor to Regulate SVZ Lineage Progression, Cell Stem Cell, vol.11, issue.2, pp.220-230, 2012.
DOI : 10.1016/j.stem.2012.06.016

H. Bramlett and W. Dietrich, Pathophysiology of cerebral ischemia and brain trauma: Similarities and differences, Journal of Cerebral Blood Flow & Metabolism, vol.784, pp.133-150, 2004.
DOI : 10.1097/01.WCB.0000111614.19196.04

E. Günther and L. Walter, The major histocompatibility complex of the rat (Rattusnorvegicus), pp.520-542, 2001.

I. Ethell and D. Ethell, Matrix metalloproteinases in brain development and remodeling: Synaptic functions and targets, Journal of Neuroscience Research, vol.275, issue.13, pp.2813-2823, 2007.
DOI : 10.1002/jnr.21273

E. Ezratty, C. Bertaux, E. Marcantonio, and G. Gundersen, Clathrin mediates integrin endocytosis for focal adhesion disassembly in migrating cells, The Journal of Cell Biology, vol.112, issue.5, pp.733-747, 2009.
DOI : 10.1128/MCB.01147-05