, Corresponding author J. Gallego: INSERM, U676, Hôpital Robert Debré, p.75019, 2012.

A. I. Abbreviations,

, arbitrary unit; pFRG, parafacial respiratory group; 5-HIAA, 5-hydroxyindoleacetic acid; HVD, hypoxic ventilatory decline; IS, irregularity score

M. and M. Oxidase-a;-pb,

R. Rmg and . Magnus,

. Rob,

. Rpa,

R. Rpn and . Pontis,

, RRG, respiratory rhythm generator

A. C. Anderica-romero, I. G. Gonzalez-herrera, A. Santamaria, and J. Pedrazachaverri, Cullin 3 as a novel target in diverse pathologies, Redox Biol, vol.1, pp.366-372, 2013.

B. K. Boh, M. Y. Ng, Y. C. Leck, B. Shaw, J. Long et al.,

R. Searle, T. Layfield, and . Hagen, Inhibition of cullin RING ligases by cycle inhibiting factor: evidence for interference with Nedd8-induced conformational control, J Mol Biol, vol.413, issue.2, pp.430-437, 2011.

N. C. Chang, M. Nguyen, M. Germain, and G. C. Shore, Antagonism of Beclin 1-dependent autophagy by BCL-2 at the endoplasmic reticulum requires NAF-1, EMBO J, vol.29, issue.3, pp.606-618, 2010.

Y. Chen and L. Yu, Autophagic lysosome reformation, Exp Cell Res, vol.319, issue.2, pp.142-146, 2013.

K. G. Commons, Locally collateralizing glutamate neurons in the dorsal raphe nucleus responsive to substance P contain vesicular glutamate transporter 3 (VGLUT3), J Chem Neuroanat, vol.38, issue.4, pp.273-281, 2009.

S. El-mestikawy, A. Wallen-mackenzie, G. M. Fortin, L. Descarries, and L. E. Trudeau, From glutamate co-release to vesicular synergy: vesicular glutamate transporters, Nat Rev Neurosci, vol.12, issue.4, pp.204-216, 2011.

W. Feng, S. Huang, H. Wu, and M. Zhang, Molecular basis of Bcl-xL's target recognition versatility revealed by the structure of Bcl-xL in complex with the BH3 domain of Beclin-1, J Mol Biol, vol.372, issue.1, pp.223-235, 2007.

G. R. Fraser, Profound Childhood Deafness, J Med Genet, vol.1, issue.2, pp.118-151, 1964.

C. Gras, B. Amilhon, E. M. Lepicard, O. Poirel, J. Vinatier et al.,

M. R. Tzavara, G. G. Wade, N. Nomikos, F. Hanoun, M. L. Saurini et al., The vesicular glutamate transporter VGLUT3 synergizes striatal acetylcholine tone, Nat Neurosci, vol.11, issue.3, pp.292-300, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00263930

C. Gras, E. Herzog, G. C. Bellenchi, V. Bernard, P. Ravassard et al., A third vesicular glutamate transporter expressed by cholinergic and serotoninergic neurons, J Neurosci, vol.22, issue.13, pp.5442-5451, 2002.

C. C. Greene, P. M. Mcmillan, S. E. Barker, P. Kurnool, M. I. Lomax et al., DFNA25, a novel locus for dominant nonsyndromic hereditary hearing impairment, maps to 12q21-24, Am J Hum Genet, vol.68, issue.1, pp.254-260, 2001.

M. A. Herman, F. Ackermann, T. Trimbuch, and C. Rosenmund, Vesicular glutamate transporter expression level affects synaptic vesicle release probability at hippocampal synapses in culture, J Neurosci, vol.34, issue.35, pp.11781-11791, 2014.

E. Herzog, F. Nadrigny, K. Silm, C. Biesemann, I. Helling et al.,

U. V. Schwartzmann, S. E. Nagerl, J. Mestikawy, F. Rhee, N. Kirchhoff et al., In vivo imaging of intersynaptic vesicle exchange using VGLUT1 Venus knock-in mice, J Neurosci, vol.31, issue.43, pp.15544-15559, 2011.

A. J. Hudspeth, Extracellular current flow and the site of transduction by vertebrate hair cells, J Neurosci, vol.2, issue.1, pp.1-10, 1982.

B. Kachar, A. Battaglia, and J. Fex, Compartmentalized vesicular traffic around the hair cell cuticular plate, Hear Res, vol.107, issue.1-2, pp.102-112, 1997.

Y. R. Lee, W. C. Yuan, H. C. Ho, C. H. Chen, H. M. Shih et al., The Cullin 3 substrate adaptor KLHL20 mediates DAPK ubiquitination to control interferon responses, EMBO J, vol.29, issue.10, pp.1748-1761, 2010.

R. T. Marquez and L. Xu, Bcl-2:Beclin 1 complex: multiple, mechanisms regulating autophagy/apoptosis toggle switch, Am J Cancer Res, vol.2, issue.2, pp.214-221, 2012.

J. Menardo, Y. Tang, S. Ladrech, M. Lenoir, F. Casas et al., Oxidative stress, inflammation, and autophagic stress as the key mechanisms of premature age-related hearing loss in SAMP8 mouse Cochlea, Antioxid Redox Signal, vol.16, issue.3, pp.263-274, 2012.

M. Morin, K. E. Bryan, F. Mayo-merino, R. Goodyear, A. Mencia et al.,

I. Castillo, J. M. Cabalka, G. Richardson, F. Moreno, P. A. Rubenstein et al., In vivo and in vitro effects of two novel gamma-actin (ACTG1) mutations that cause DFNA20/26 hearing impairment, Hum Mol Genet, vol.18, issue.16, pp.3075-3089, 2009.

A. Oberstein, P. D. Jeffrey, and Y. Shi, Crystal structure of the Bcl-XL-Beclin 1 peptide complex: Beclin 1 is a novel BH3-only protein, J Biol Chem, vol.282, issue.17, pp.13123-13132, 2007.

Y. Ohsumi, Historical landmarks of autophagy research, Cell Res, vol.24, issue.1, pp.9-23, 2014.

J. Qi, S. Zhang, H. L. Wang, H. Wang, J. De-jesus-aceves et al.,

R. Lupica, R. P. Seal, and M. Morales, A glutamatergic reward input from the dorsal raphe to ventral tegmental area dopamine neurons, Nat Commun, vol.5, p.5390, 2014.

J. Ruel, S. Emery, R. Nouvian, T. Bersot, B. Amilhon et al., Impairment of SLC17A8 encoding vesicular glutamate transporter-3, VGLUT3, underlies nonsyndromic deafness DFNA25 and inner hair cell dysfunction in null mice, Am J Hum Genet, vol.83, issue.2, pp.278-292, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00349467

D. Y. Sakae, F. Marti, S. Lecca, F. Vorspan, E. Martin-garcia et al., The absence of VGLUT3 predisposes to cocaine abuse by increasing dopamine and glutamate signaling in the nucleus accumbens, Mol Psychiatry, vol.20, issue.11, pp.1448-1459, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01536677

M. K. Schafer, H. Varoqui, N. Defamie, E. Weihe, and J. D. Erickson, Molecular cloning and functional identification of mouse vesicular glutamate transporter 3 and its expression in subsets of novel excitatory neurons, J Biol Chem, vol.277, issue.52, pp.50734-50748, 2002.

R. P. Seal, O. Akil, E. Yi, C. M. Weber, L. Grant et al.,

E. Noebels, L. R. Glowatzki, R. H. Lustig, and . Edwards, Sensorineural deafness and seizures in mice lacking vesicular glutamate transporter 3, Neuron, vol.57, issue.2, pp.263-275, 2008.

G. Sendin, A. V. Bulankina, D. Riedel, and T. Moser, Maturation of ribbon synapses in hair cells is driven by thyroid hormone, J Neurosci, vol.27, issue.12, pp.3163-3173, 2007.

H. M. Shen and P. Codogno, Autophagic cell death: Loch Ness monster or endangered species?, Autophagy, vol.7, issue.5, pp.457-465, 2011.

L. Siksou, K. Silm, C. Biesemann, R. B. Nehring, S. M. Wojcik et al.,

S. Mestikawy, E. Marty, and . Herzog, A role for vesicular glutamate transporter 1 in synaptic vesicle clustering and mobility, Eur J Neurosci, vol.37, issue.10, pp.1631-1642, 2013.

S. Takamori, P. Malherbe, C. Broger, R. Jahn-;-van-wijk, E. et al., Molecular cloning and functional characterization of human vesicular glutamate transporter 3, EMBO Rep, vol.3, issue.8, pp.798-803, 2002.

F. P. Cremers, H. Cremers, and . Kremer, A mutation in the gamma actin 1 (ACTG1) gene causes autosomal dominant hearing loss (DFNA20/26), J Med Genet, vol.40, issue.12, pp.879-884, 2003.

E. Vigneault, O. Poirel, M. Riad, J. Prud'homme, S. Dumas et al., Distribution of vesicular glutamate transporters in the human brain, Front Neuroanat, vol.9, p.23, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01221681

M. C. Weston, R. B. Nehring, S. M. Wojcik, and C. Rosenmund, Interplay between VGLUT isoforms and endophilin A1 regulates neurotransmitter release and shortterm plasticity, Neuron, vol.69, issue.6, pp.1147-1159, 2011.

E. Wirawan, T. Vanden-berghe, S. Lippens, P. Agostinis, and P. Vandenabeele, Autophagy: for better or for worse, Cell Res, vol.22, issue.1, pp.43-61, 2012.

E. Zalckvar, H. Berissi, M. Eisenstein, and A. Kimchi, Phosphorylation of Beclin 1 by DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL, Autophagy, vol.5, issue.5, pp.720-722, 2009.

M. Zhu, T. Yang, S. Wei, A. T. Dewan, R. J. Morell et al.,

R. J. Leal, K. H. Smith, and . Friderici, rose circles), 1000 (red circles) and 2000 (black circles) µM of LSP5-2157. All points represent mean ± S.E. values calculated from seven animals. Note almost no effect of LSP52157 on CM and SP. By contrast, LSP5-2157 caused a drastic reduction in CAP amplitude and an increase in N1 latency. E: Dose-response curves obtained with LSP5-2157. The dose-responses were fitted to a curve using a non-linear least-square logistic equation. The IC50 value, Am J Hum Genet, vol.73, issue.5, pp.1082-1091, 2003.

, SAMP8-EE et SAMR1 valeur de p = 0.0004, comparaison SAMP8-NE et SAMR1 valeur de p =0.08)

, Ces différences disparaissent à 9 mois pour la latence (test de Fisher, comparaison des groupes SAMP8-EE et SAMP8-NE valeur de p = 0.28, comparaison SAMP8-EE et SAMR1 valeur de p = 0.59, comparaison SAMP8-NE et SAMR1 valeur de p =0.12), le temps d'immobilité (test de Fisher, comparaison des groupes SAMP8-EE et SAMP8-NE valeur de p = 0.16, comparaison SAMP8-EE et SAMR1 valeur de p = 0.9, comparaison SAMP8NE et SAMR1 valeur de p = 0.12) et le nombre de périodes d'immobilité

, SAMP8-EE et SAMR1 valeur de p = 0.62, comparaison SAMP8-NE et SAMR1 valeur de p =, vol.14, p.0

V. Abadie, J. Champagnat, and G. Fortin, Branchiomotor activities in mouse embryo, Neuroreport, vol.11, issue.1, pp.141-145, 2000.
URL : https://hal.archives-ouvertes.fr/hal-00122695

J. M. Adams and S. Cory, The Bcl-2 apoptotic switch in cancer development and therapy, Oncogene, vol.26, issue.9, pp.1324-1337, 2007.

Y. Aihara, H. Mashima, H. Onda, S. Hisano, H. Kasuya et al., Molecular cloning of a novel braintype Na(+)-dependent inorganic phosphate cotransporter, J Neurochem, vol.74, issue.6, p.2622, 2000.

N. Alenina, D. Kikic, M. Todiras, V. Mosienko, F. Qadri et al.,

R. Vilianovitch, K. Sohr, H. Tenner, M. Hortnagl, and . Bader, Growth retardation and altered autonomic control in mice lacking brain serotonin, Proc Natl Acad Sci U S A, vol.106, issue.25, pp.10332-10337, 2009.

A. E. Allain, P. Meyrand, and P. Branchereau, , 2005.

, GABAergic cell population are controlled by the serotonin (5-HT) system: implication of 5-HT1 receptor family, J Neurosci, vol.25, issue.38, pp.8714-8724

J. Almqvist, Y. Huang, A. Laaksonen, D. N. Wang, and S. Hovmoller, Docking and homology modeling explain inhibition of the human vesicular glutamate transporters, Protein Sci, vol.16, issue.9, pp.1819-1829, 2007.

D. C. Altieri, Survivin and IAP proteins in cell-death mechanisms, Biochem J, vol.430, issue.2, pp.199-205, 2010.

O. Alvarez-garcia, I. Vega-naredo, V. Sierra, B. Caballero, C. Tomas-zapico et al.,

J. J. Camins, M. Garcia, A. Pallas, and . Coto-montes, Elevated oxidative stress in the brain of senescence-accelerated mice at 5 months of age, Biogerontology, vol.7, issue.1, p.43, 2006.

T. Amano, H. Nakanishi, M. Oka, and K. Yamamoto, Increased expression of cathepsins E and D in reactive microglial cells associated with spongiform degeneration in the brain stem of senescence-accelerated mouse, Exp Neurol, vol.136, issue.2, pp.171-182, 1995.

, DSM-V. Diagnostic and statistical manual of mental disorders, American Psychiatric Association, 2013.

J. Amiel, V. Dubreuil, N. Ramanantsoa, G. Fortin, J. Gallego et al., PHOX2B in respiratory control: lessons from congenital central hypoventilation syndrome and its mouse models, Respir Physiol Neurobiol, vol.168, issue.1-2, pp.125-132, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00444550

B. Amilhon, E. Lepicard, T. Renoir, R. Mongeau, D. Popa et al.,

M. Gardier, J. Gallego, M. Hamon, L. Lanfumey, B. Gasnier et al., VGLUT3 (vesicular glutamate transporter type 3) contribution to the regulation of serotonergic transmission and anxiety, J Neurosci, vol.30, issue.6, pp.2198-2210, 2010.

L. Bai, H. Xu, J. F. Collins, and F. K. Ghishan, Molecular and functional analysis of a novel neuronal vesicular glutamate transporter, J Biol Chem, vol.276, 2001.

B. Bayram, S. Nikolai, P. Huebbe, B. Ozcelik, S. Grimm et al.,

. Rimbach, Biomarkers of oxidative stress, antioxidant defence and inflammation are altered in the senescence-accelerated mouse prone 8, Age (Dordr), vol.35, issue.4, p.1205, 2013.

E. E. Bellocchio, H. Hu, A. Pohorille, J. Chan, V. M. Pickel et al., , 1998.

, The localization of the brain-specific inorganic phosphate transporter suggests a specific presynaptic role in glutamatergic transmission, J Neurosci, vol.18, issue.21, pp.8648-8659

E. E. Bellocchio, R. J. Reimer, R. T. Fremeau, J. , and R. H. Edwards, Uptake of glutamate into synaptic vesicles by an inorganic phosphate transporter, Science, vol.289, issue.5481, pp.957-960, 2000.

J. Belmin and C. Sebban, Physiologie du vieillissement, Gérontologie pour le praticien, pp.8-20, 2005.

C. Bernard, Leçons sur les phénomènes de la vie communs aux animaux et aux végétaux, tome premier, J.-B. Baillière et fils, seconde édition, Cell, vol.131, issue.6, pp.1137-1148, 1878.

N. Berube-carriere, M. Riad, G. Bo, D. Levesque, L. E. Trudeau et al., The dual dopamine-glutamate phenotype of growing mesencephalic neurons regresses in mature rat brain, J Comp Neurol, vol.517, issue.6, pp.873-891, 2009.

S. K. Bhutia, S. Mukhopadhyay, N. Sinha, D. N. Das, P. K. Panda et al., Autophagy: cancer's friend or foe?, Adv Cancer Res, vol.118, pp.61-95, 2013.

L. M. Bierer, V. Haroutunian, S. Gabriel, P. J. Knott, L. S. Carlin et al.,

J. Perl, P. Schmeidler, K. L. Kanof, and . Davis, Neurochemical correlates of dementia severity in Alzheimer's disease: relative importance of the cholinergic deficits, J Neurochem, vol.64, issue.2, pp.749-760, 1995.

B. Blanchi, L. M. Kelly, J. C. Viemari, I. Lafon, H. Burnet et al.,

T. Daniel, G. Graf, M. H. Hilaire, and . Sieweke, MafB deficiency causes defective respiratory rhythmogenesis and fatal central apnea at birth, Nat Neurosci, vol.6, issue.10, p.1091, 2003.

B. Bollen, M. Bouslama, B. Matrot, Y. Rotrou, G. Vardon et al.,

R. D'hooge and J. Gallego, Cold stimulates the behavioral response to hypoxia in newborn mice, Am J Physiol Regul Integr Comp Physiol, vol.296, issue.5, pp.1503-1511, 2009.

D. Bouccara, , 2005.

J. L. Boulland, T. Qureshi, R. P. Seal, A. Rafiki, V. Gundersen et al.,

R. H. Fremeau, J. Edwards, F. A. Storm-mathisen, and . Chaudhry, Expression of the vesicular glutamate transporters during development indicates the widespread corelease of multiple neurotransmitters, J Comp Neurol, vol.480, issue.3, pp.264-280, 2004.

J. Bouvier, M. Thoby-brisson, N. Renier, V. Dubreuil, J. Ericson et al.,

A. Pierani, G. Chedotal, and . Fortin, Hindbrain interneurons and axon guidance signaling critical for breathing, Nat Neurosci, vol.13, issue.9, pp.1066-1074, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00517787

E. J. Bowman, A. Siebers, and K. Altendorf, Bafilomycins: a class of inhibitors of membrane ATPases from microorganisms, animal cells, and plant cells, Proc Natl Acad Sci U S A, vol.85, issue.21, pp.7972-7976, 1988.

H. F. Bradford and H. K. Ward, On glutaminase activity in mammalian synaptosomes, Brain Res, vol.110, issue.1, pp.115-125, 1976.

H. Bras, S. P. Gaytan, P. Portalier, S. Zanella, R. Pasaro et al., Prenatal activation of 5-HT2A receptor induces expression of 5-HT1B receptor in phrenic motoneurons and alters the organization of their premotor network in newborn mice, Eur J Neurosci, vol.28, issue.6, pp.1097-1107, 2008.

L. Buée and C. A. Maurage, Le vieillissement : des molecules, des cellules et des structures cérébrales en involution, Neuropyshcologie du vieillissement nomal et pathologique, Ed Masson, pp.11-26, 2008.

D. A. Butterfield and C. B. Pocernich, The glutamatergic system and, 2003.

, Alzheimer's disease: therapeutic implications, CNS Drugs, vol.17, issue.9, pp.641-652

D. A. Butterfield and H. F. Poon, The senescence-accelerated prone mouse (SAMP8): a model of age-related cognitive decline with relevance to alterations of the gene expression and protein abnormalities in Alzheimer's disease, Exp Gerontol, vol.40, issue.10, pp.774-783, 2005.

V. A. Campbell, J. E. Crews, D. G. Moriarty, M. M. Zack, and D. K. Blackman, , 1999.

, Surveillance for sensory impairment, activity limitation, and health-related quality of life among older adults-United States, MMWR CDC Surveill Summ, vol.48, issue.8, pp.131-156, 1993.

J. Campisi, Aging, cellular senescence, and cancer, Annu Rev Physiol, vol.75, pp.685-705, 2013.

A. M. Canudas, J. Gutierrez-cuesta, M. I. Rodriguez, D. Acuna-castroviejo, and F. ,

A. Sureda, M. Camins, and . Pallas, Hyperphosphorylation of microtubuleassociated protein tau in senescence-accelerated mouse (SAM), Mech Ageing Dev, vol.126, issue.12, pp.1300-1304, 2005.

S. Carson, M. S. Mcdonagh, and K. Peterson, A systematic review of the efficacy and safety of atypical antipsychotics in patients with psychological and behavioral symptoms of dementia, J Am Geriatr Soc, vol.54, issue.2, pp.354-361, 2006.

X. Caubit, M. Thoby-brisson, N. Voituron, P. Filippi, M. Bevengut et al.,

G. Zanella, G. Fortin, L. Hilaire, and . Fasano, Teashirt 3 regulates development of neurons involved in both respiratory rhythm and airflow control, J Neurosci, vol.30, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00555206

V. J. Cerpa, L. Aylwin-mde, S. Beltran-castillo, E. U. Bravo, I. R. Llona et al., The Alteration of Neonatal Raphe Neurons by PrenatalPerinatal Nicotine. Meaning for Sudden Infant Death Syndrome, Am J Respir Cell Mol Biol, vol.53, issue.4, pp.489-499, 2015.

T. Chandra and K. Kirschner, Chromosome organisation during ageing and senescence, Curr Opin Cell Biol, vol.40, pp.161-167, 2016.

N. C. Chang, M. Nguyen, M. Germain, and G. C. Shore, Antagonism of Beclin 1dependent autophagy by BCL-2 at the endoplasmic reticulum requires NAF-1, EMBO J, vol.29, issue.3, pp.606-618, 2010.

G. H. Chen, Y. J. Wang, X. M. Wang, and J. N. Zhou, Accelerated senescence prone mouse-8 shows early onset of deficits in spatial learning and memory in the radial six-arm water maze, Physiol Behav, vol.82, issue.5, pp.883-890, 2004.

X. Chen, W. Li, J. Ren, D. Huang, W. T. He et al., Translocation of mixed lineage kinase domain-like protein to plasma membrane leads to necrotic cell death, Cell Res, vol.24, issue.1, pp.105-121, 2014.

Y. Chen, Z. Yang, M. Meng, Y. Zhao, N. Dong et al.,

. Shao, Cullin mediates degradation of RhoA through evolutionarily conserved BTB adaptors to control actin cytoskeleton structure and cell movement, Mol Cell, vol.35, issue.6, pp.841-855, 2009.

Y. Chen and L. Yu, Autophagic lysosome reformation, Exp Cell Res, vol.319, issue.2, 2013.

E. H. Cheng, M. C. Wei, S. Weiler, R. A. Flavell, T. W. Mak et al.,

. Korsmeyer, BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX-and BAK-mediated mitochondrial apoptosis, Mol Cell, vol.8, issue.3, pp.705-711, 2001.

X. R. Cheng, Y. Yang, W. X. Zhou, and Y. X. Zhang, Expression of VGLUTs contributes to degeneration and acquisition of learning and memory, Neurobiol Learn Mem, vol.95, issue.3, pp.361-375, 2011.

B. G. Childs, D. J. Baker, T. Wijshake, C. A. Conover, J. Campisi et al., Senescent intimal foam cells are deleterious at all stages of atherosclerosis, Science, vol.354, issue.6311, pp.472-477, 2016.

E. D. Christensen, J. Berger, M. M. Alashari, H. Coon, C. Robison et al., Sudden infant death "syndrome"-Insights and future directions from a Utah population database analysis, 2016.

, Am J Med Genet A

H. R. Chung, M. Vakil, M. Munroe, A. Parikh, B. M. Meador et al.,

K. R. Woods, M. D. Wilund, and . Boppart, The Impact of Exercise on StatinAssociated Skeletal Muscle Myopathy, PLoS One, vol.11, issue.12, p.168065, 2016.

A. Clegg, J. Young, S. Iliffe, M. O. Rikkert, and K. Rockwood, Frailty in elderly people, Lancet, vol.381, issue.9868, pp.752-762, 2013.

T. J. Collier, J. G. Greene, D. L. Felten, S. Y. Stevens, and K. S. Collier, Reduced cortical noradrenergic neurotransmission is associated with increased neophobia and impaired spatial memory in aged rats, Neurobiol Aging, vol.25, issue.2, pp.209-221, 2004.

G. L. Collingridge, S. Peineau, J. G. Howland, and Y. T. Wang, Long-term depression in the CNS, Nat Rev Neurosci, vol.11, issue.7, pp.459-473, 2010.

C. Costantini, H. Scrable, and L. Puglielli, An aging pathway controls the TrkA to p75NTR receptor switch and amyloid beta-peptide generation, EMBO J, vol.25, issue.9, 2006.

P. J. Coucke, P. Van-hauwe, P. M. Kelley, H. Kunst, I. Schatteman et al.,

R. J. Meyers, M. Ensink, F. Verstreken, H. Declau, K. Marres et al.,

R. J. Mcguirt, C. W. Smith, P. Cremers, P. J. Van-de-heyning, S. D. Willems et al.,

V. Camp, Mutations in the KCNQ4 gene are responsible for autosomal dominant deafness in four DFNA2 families, Hum Mol Genet, vol.8, issue.7, pp.1321-1328, 1999.

P. Courville, M. Quick, and R. J. Reimer, Structure-function studies of the SLC17 transporter sialin identify crucial residues and substrate-induced conformational changes, J Biol Chem, vol.285, issue.25, pp.19316-19323, 2010.

K. J. Cruickshanks, T. L. Wiley, T. S. Tweed, B. E. Klein, R. Klein et al., Prevalence of hearing loss in older adults in Beaver Dam, Wisconsin. The Epidemiology of Hearing Loss Study, Am J Epidemiol, vol.148, issue.9, pp.879-886, 1998.

J. L. Cummings, Alzheimer's disease, N Engl J Med, vol.351, issue.1, pp.56-67, 2004.
URL : https://hal.archives-ouvertes.fr/hal-01579097

K. J. Cummings, Aspiring to inspire-serotonin, the laryngeal chemoreflex and the sudden infant death syndrome, Exp Physiol, vol.101, issue.7, p.790, 2016.

K. J. Cummings, K. G. Commons, J. C. Hewitt, J. A. Daubenspeck, A. Li et al., Failed heart rate recovery at a critical age in 5-HT-deficient mice exposed to episodic anoxia: implications for SIDS, J Appl Physiol, vol.111, issue.3, p.825, 1985.

G. Dal-bo, N. Berube-carriere, J. A. Mendez, D. Leo, M. Riad et al.,

L. E. Levesque and . Trudeau, Enhanced glutamatergic phenotype of mesencephalic dopamine neurons after neonatal 6-hydroxydopamine lesion, Neuroscience, vol.156, issue.1, pp.59-70, 2008.

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

R. W. Daniels, C. A. Collins, K. Chen, M. V. Gelfand, D. E. Featherstone et al.,

. Diantonio, A single vesicular glutamate transporter is sufficient to fill a synaptic vesicle, Neuron, vol.49, issue.1, pp.11-16, 2006.

S. Daumas, J. Sandin, K. S. Chen, D. Kobayashi, J. Tulloch et al.,

G. Morris, Faster forgetting contributes to impaired spatial memory in the PDAPP mouse: deficit in memory retrieval associated with increased sensitivity to interference?, Learn Mem, vol.15, issue.9, pp.625-632, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02351279

Z. Davanipour, N. M. Lu, M. Lichtenstein, and K. S. Markides, Hearing problems in Mexican American elderly, Am J Otol, vol.21, issue.2, pp.168-172, 2000.

L. M. Dejean, S. Martinez-caballero, L. Guo, C. Hughes, O. Teijido et al.,

S. J. Korsmeyer, B. Antonsson, E. A. Jonas, and K. W. Kinnally, Oligomeric Bax is a component of the putative cytochrome c release channel MAC, mitochondrial apoptosisinduced channel, Mol Biol Cell, vol.16, issue.5, pp.2424-2432, 2005.

J. Del-valle, S. Bayod, A. Camins, C. Beas-zarate, D. A. Velazquez-zamora et al., Dendritic spine abnormalities in hippocampal CA1, 2012.

, Alzheimer's disease, J Alzheimers Dis, vol.32, issue.1, pp.233-240

D. Valle, J. , J. Duran-vilaregut, G. Manich, A. Camins et al.,

. Pelegri, Time-course of blood-brain barrier disruption in senescenceaccelerated mouse prone 8 (SAMP8) mice, Int J Dev Neurosci, vol.27, issue.1, pp.47-52, 2009.

D. Valle, J. , J. Duran-vilaregut, G. Manich, G. Casadesus et al.,

C. Pallas, J. Pelegri, and . Vilaplana, Early amyloid accumulation in the hippocampus of SAMP8 mice, J Alzheimers Dis, vol.19, issue.4, pp.1303-1315, 2010.

J. Del-valle, J. Duran-vilaregut, G. Manich, M. Pallas, A. Camins et al.,

. Pelegri, Cerebral amyloid angiopathy, blood-brain barrier disruption and amyloid accumulation in SAMP8 mice, Neurodegener Dis, vol.8, issue.6, pp.421-429, 2011.

L. Deng, J. Yao, C. Fang, N. Dong, B. Luscher et al., Sequential postsynaptic maturation governs the temporal order of GABAergic and glutamatergic synaptogenesis in rat embryonic cultures, J Neurosci, vol.27, issue.40, pp.10860-10869, 2007.

D. Denton, S. Nicolson, and S. Kumar, Cell death by autophagy: facts and apparent artefacts, Cell Death Differ, vol.19, issue.1, pp.87-95, 2012.

M. Dobarro, L. Orejana, N. Aguirre, and M. J. Ramirez, Propranolol restores cognitive deficits and improves amyloid and Tau pathologies in a senescenceaccelerated mouse model, Neuropharmacology, vol.64, pp.137-144, 2013.

W. T. Donnelly, D. Bartlett, J. , and J. C. Leiter, Serotonin in the solitary tract nucleus shortens the laryngeal chemoreflex in anaesthetized neonatal rats, Exp Physiol, vol.101, issue.7, pp.946-961, 2016.

P. Duan and L. F. Bonewald, The role of the wnt/beta-catenin signaling pathway in formation and maintenance of bone and teeth, Int J Biochem Cell Biol, vol.77, pp.23-29, 2016.

K. Dujardin and P. Lemaire, Neurophysiologie du vieillissement normal et pathologique, 2008.

J. R. Duncan, D. S. Paterson, J. M. Hoffman, D. J. Mokler, N. S. Borenstein et al.,

H. F. Belliveau, E. A. Krous, C. Haas, E. E. Stanley, F. L. Nattie et al.,

. Kinney, Brainstem serotonergic deficiency in sudden infant death syndrome, JAMA, vol.303, issue.5, pp.430-437, 2010.

R. H. Edwards, The neurotransmitter cycle and quantal size, Neuron, vol.55, issue.6, pp.835-858, 2007.

L. E. Eiden, The vesicular neurotransmitter transporters: current perspectives and future prospects, FASEB J, vol.14, issue.15, pp.2396-2400, 2000.

S. El-mestikawy, A. Wallen-mackenzie, G. M. Fortin, L. Descarries, and L. E. Trudeau, From glutamate co-release to vesicular synergy: vesicular glutamate transporters, Nat Rev Neurosci, vol.12, issue.4, pp.204-216, 2011.

M. Elgendy, C. Sheridan, G. Brumatti, and S. J. Martin, Oncogenic Rasinduced expression of Noxa and Beclin-1 promotes autophagic cell death and limits clonogenic survival, Mol Cell, vol.42, issue.1, pp.23-35, 2011.

D. W. Ellison, M. F. Beal, M. F. Mazurek, E. D. Bird, and J. B. Martin, A postmortem study of amino acid neurotransmitters in Alzheimer's disease, Ann Neurol, vol.20, issue.5, pp.616-621, 1986.

J. T. Erickson, G. Shafer, M. D. Rossetti, C. G. Wilson, and E. S. Deneris, Arrest of 5HT neuron differentiation delays respiratory maturation and impairs neonatal homeostatic responses to environmental challenges, Respir Physiol Neurobiol, vol.159, issue.1, pp.85-101, 2007.

R. Eskes, S. Desagher, B. Antonsson, and J. C. Martinou, Bid induces the oligomerization and insertion of Bax into the outer mitochondrial membrane, Mol Cell Biol, vol.20, issue.3, pp.929-935, 2000.

M. Eybalin, Neurotransmitters and neuromodulators of the mammalian cochlea, Physiol Rev, vol.73, issue.2, pp.309-373, 1993.

S. Farley, S. Dumas, S. E. Mestikawy, and B. Giros, Increased expression of the Vesicular Glutamate Transporter-1 (VGLUT1) in the prefrontal cortex correlates with differential vulnerability to chronic stress in various mouse strains: effects of fluoxetine and MK-801, Neuropharmacology, vol.62, issue.1, pp.503-517, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01542336

F. C. Favre-besse, O. Poirel, T. Bersot, E. Kim-grellier, S. Daumas et al.,

C. Acher and N. Pietrancosta, Design, synthesis and biological evaluation of small-azo-dyes as potent Vesicular Glutamate Transporters inhibitors, Eur J Med Chem, vol.78, pp.236-247, 2014.

W. Feng, S. Huang, H. Wu, and M. Zhang, Molecular basis of Bcl-xL's target recognition versatility revealed by the structure of Bcl-xL in complex with the BH3 domain of Beclin-1, J Mol Biol, vol.372, issue.1, pp.223-235, 2007.

M. Fenton, H. L. Huang, Y. Hong, E. Hawe, D. J. Kurz et al., , 2004.

, Early atherogenesis in senescence-accelerated mice, Exp Gerontol, vol.39, issue.1, pp.115-122

J. F. Flood, S. A. Farr, F. E. Kaiser, and J. E. Morley, Age-related impairment in learning but not memory in SAMP8 female mice, Pharmacol Biochem Behav, vol.50, issue.4, pp.661-664, 1995.

J. F. Flood, F. J. Harris, and J. E. Morley, Age-related changes in hippocampal drug facilitation of memory processing in SAMP8 mice, Neurobiol Aging, vol.17, issue.1, pp.15-24, 1996.

J. F. Flood and J. E. Morley, Early onset of age-related impairment of aversive and appetitive learning in the SAM-P/8 mouse, J Gerontol, vol.47, issue.2, pp.52-59, 1992.

J. F. Flood and J. E. Morley, Learning and memory in the SAMP8 mouse, 1998.

, Neurosci Biobehav Rev, vol.22, issue.1, pp.1-20

J. F. Flood, J. E. Morley, and M. L. Reginna, Age-related changes in the pharmacological improvement of retention in senescence accelerated mouse (SAM), Neurobiol Aging, vol.14, issue.2, pp.159-166, 1993.

R. T. Fremeau, J. Jr, T. Burman, C. H. Qureshi, J. Tran et al.,

D. R. Sulzer, J. Copenhagen, R. J. Storm-mathisen, F. A. Reimer, and R. Chaudhry,

. Edwards, The identification of vesicular glutamate transporter 3 suggests novel modes of signaling by glutamate, Proc Natl Acad Sci U S A, vol.99, issue.22, pp.14488-14493, 2002.

R. T. Fremeau, M. D. Jr, I. Troyer, G. O. Pahner, C. H. Nygaard et al.,

D. Bellocchio, J. Fortin, R. H. Storm-mathisen, and . Edwards, The expression of vesicular glutamate transporters defines two classes of excitatory synapse, Neuron, vol.31, issue.2, pp.247-260, 2001.

L. P. Fried, C. M. Tangen, J. Walston, A. B. Newman, C. Hirsch et al.,

R. Seeman, W. J. Tracy, G. Kop, M. A. Burke, G. Mcburnie et al., Frailty in older adults: evidence for a phenotype, Health Study Collaborative Research, 2001.

, Gerontol A Biol Sci Med Sci, vol.56, issue.3, pp.146-156

Y. Fujibayashi, S. Yamamoto, A. Waki, J. Konishi, and Y. Yonekura, Increased mitochondrial DNA deletion in the brain of SAMP8, a mouse model for spontaneous oxidative stress brain, Neurosci Lett, vol.254, issue.2, pp.109-112, 1998.

F. Fujiyama, T. Furuta, and T. Kaneko, Immunocytochemical localization of candidates for vesicular glutamate transporters in the rat cerebral cortex, J Comp Neurol, vol.435, issue.3, pp.379-387, 2001.

L. Galluzzi, J. M. Vitale, E. S. Abrams, E. H. Alnemri, M. V. Baehrecke et al.,

O. Hengartner, R. A. Kepp, S. Knight, S. A. Kumar, X. Lipton et al.,

G. Mehlen, M. E. Nunez, M. Peter, D. C. Piacentini, Y. Rubinsztein et al.,

E. Vandenabeele, J. White, B. Yuan, G. Zhivotovsky, G. Melino et al., , 2012.

, Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012, Cell Death Differ, vol.19, issue.1, pp.107-120

G. Gangarossa, M. Guzman, V. F. Prado, M. A. Prado, S. Daumas et al., Role of the atypical vesicular glutamate transporter VGLUT3 in lDOPA-induced dyskinesia, Neurobiol Dis, vol.87, pp.69-79, 2016.

C. J. Garwood, L. E. Ratcliffe, J. E. Simpson, P. R. Heath, P. G. Ince et al., Review: Astrocytes in Alzheimer's disease and other age, 2016.

, a supporting player with a central role, Neuropathol Appl Neurobiol

B. Gasnier, The loading of neurotransmitters into synaptic vesicles, Biochimie, vol.82, issue.4, pp.327-337, 2000.

P. Gaspar, O. Cases, and L. Maroteaux, The developmental role of serotonin: news from mouse molecular genetics, Nat Rev Neurosci, vol.4, issue.12, pp.1002-1012, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01274960

C. L. Gatto and K. Broadie, Genetic controls balancing excitatory and inhibitory synaptogenesis in neurodevelopmental disorder models, Front Synaptic Neurosci, vol.2, p.4, 2010.

E. Georgakopoulou, K. Evangelou, S. Havaki, P. Townsend, P. Kanavaros et al.,

. Gorgoulis, Apoptosis or senescence? Which exit route do epithelial cells and fibroblasts preferentially follow?, Mech Ageing Dev, vol.156, pp.17-24, 2016.

P. Gil-loyzaga and R. Pujol, Synaptophysin in the developing cochlea, Int J, 1988.

, Dev Neurosci, vol.6, issue.2, pp.155-160

D. C. Gillespie, G. Kim, and K. Kandler, Inhibitory synapses in the developing auditory system are glutamatergic, Nat Neurosci, vol.8, issue.3, pp.332-338, 2005.

C. Girbovan and H. Plamondon, Environmental enrichment in female rodents: considerations in the effects on behavior and biochemical markers, Behav Brain Res, vol.253, pp.178-190, 2013.

Y. Gong, L. Liu, B. Xie, Y. Liao, E. Yang et al., Ameliorative effects of lotus seedpod proanthocyanidins on cognitive deficits and oxidative damage in senescence-accelerated mice, Behav Brain Res, vol.194, issue.1, pp.100-107, 2008.

N. Gortz, L. Lewejohann, M. Tomm, O. Ambree, K. Keyvani et al., Effects of environmental enrichment on exploration, anxiety, and memory in female TgCRND8 Alzheimer mice, Behav Brain Res, vol.191, issue.1, pp.43-48, 2008.

D. Grander, P. Kharaziha, E. Laane, K. Pokrovskaja, and T. Panaretakis, , 2009.

, Autophagy as the main means of cytotoxicity by glucocorticoids in hematological malignancies, Autophagy, vol.5, issue.8, pp.1198-1200

B. Granseth, F. K. Andersson, and S. H. Lindstrom, The initial stage of reversal learning is impaired in mice hemizygous for the vesicular glutamate transporter (VGluT1), Genes Brain Behav, vol.14, issue.6, pp.477-485, 2015.

L. Grant, E. Yi, and E. Glowatzki, Two modes of release shape the postsynaptic response at the inner hair cell ribbon synapse, J Neurosci, vol.30, issue.12, p.4210, 2010.

C. Gras, B. Amilhon, E. M. Lepicard, O. Poirel, J. Vinatier et al.,

M. R. Tzavara, G. G. Wade, N. Nomikos, F. Hanoun, M. L. Saurini et al., The vesicular glutamate transporter VGLUT3 synergizes striatal acetylcholine tone, Nat Neurosci, vol.11, issue.3, pp.292-300, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00263930

C. Gras, E. Herzog, G. C. Bellenchi, V. Bernard, P. Ravassard et al., A third vesicular glutamate transporter expressed by cholinergic and serotoninergic neurons, J Neurosci, vol.22, issue.13, pp.5442-5451, 2002.

C. Gras, J. Vinatier, B. Amilhon, A. Guerci, C. Christov et al., Developmentally regulated expression of VGLUT3 during early postnatal life, Neuropharmacology, vol.49, issue.6, pp.901-911, 2005.

F. D. Gregory, K. E. Bryan, T. Pangrsic, I. E. Calin-jageman, T. Moser et al., Harmonin inhibits presynaptic Cav1.3 Ca(2)(+) channels in mouse inner hair cells, Nat Neurosci, vol.14, issue.9, pp.1109-1111, 2011.

A. Gross, X. M. Yin, K. Wang, M. C. Wei, J. Jockel et al.,

P. Tempst and S. J. Korsmeyer, Caspase cleaved BID targets mitochondria and is required for cytochrome c release, while BCL-XL prevents this release but not tumor necrosis factor-R1/Fas death, J Biol Chem, vol.274, issue.2, pp.1156-1163, 1999.

J. Haddad and B. Langer, Médecine foetale et néonatale, 2004.

C. A. Hales, S. A. Stuart, M. H. Anderson, and E. S. Robinson, Modelling cognitive affective biases in major depressive disorder using rodents, Br J Pharmacol, vol.171, issue.20, pp.4524-4538, 2014.

H. Harati, A. Barbelivien, K. Herbeaux, M. A. Muller, M. Engeln et al., Lifelong environmental enrichment in rats: impact on emotional behavior, spatial memory vividness, and cholinergic neurons over the lifespan, Age (Dordr), vol.35, issue.4, pp.1027-1043, 2013.

J. Harris, Autophagy and cytokines, Cytokine, vol.56, issue.2, pp.140-144, 2011.

J. Hartinger and R. Jahn, An anion binding site that regulates the glutamate transporter of synaptic vesicles, J Biol Chem, vol.268, issue.31, pp.23122-23127, 1993.

L. Hayflick and P. S. Moorhead, The serial cultivation of human diploid cell strains, Exp Cell Res, vol.25, pp.585-621, 1961.

S. He, Y. Liang, F. Shao, and X. Wang, Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3mediated pathway, Proc Natl Acad Sci U S A, vol.108, issue.50, pp.20054-20059, 2011.

T. J. Hendricks, D. V. Fyodorov, L. J. Wegman, N. B. Lelutiu, E. A. Pehek et al., Pet-1 ETS gene plays a critical role in 5-HT neuron development and is required for normal anxiety-like and aggressive behavior, Neuron, vol.37, issue.2, pp.233-247, 2003.

E. Herzog, G. C. Bellenchi, C. Gras, V. Bernard, P. Ravassard et al., The existence of a second vesicular glutamate transporter specifies subpopulations of glutamatergic neurons, J Neurosci, vol.21, 2001.

E. Herzog, J. Gilchrist, C. Gras, A. Muzerelle, P. Ravassard et al., Localization of VGLUT3, the vesicular glutamate transporter type 3, in the rat brain, Neuroscience, vol.123, issue.4, pp.983-1002, 2004.

E. Herzog, M. Landry, E. Buhler, R. Bouali-benazzouz, C. Legay et al.,

P. Nagy, B. Dreyfus, S. E. Giros, and . Mestikawy, Expression of vesicular glutamate transporters, VGLUT1 and VGLUT2, in cholinergic spinal motoneurons, Eur J Neurosci, vol.20, issue.7, pp.1752-1760, 2004.

E. Herzog, F. Nadrigny, K. Silm, C. Biesemann, I. Helling et al.,

U. V. Schwartzmann, S. E. Nagerl, J. Mestikawy, F. Rhee, N. Kirchhoff et al., , 2011.

, In vivo imaging of intersynaptic vesicle exchange using VGLUT1 Venus knock-in mice, J Neurosci, vol.31, issue.43, pp.15544-15559

M. J. Higley, A. H. Gittis, I. A. Oldenburg, N. Balthasar, R. P. Seal et al.,

A. C. Lowell, B. L. Kreitzer, and . Sabatini, Cholinergic interneurons mediate fast, 2011.

, VGluT3-dependent glutamatergic transmission in the striatum, PLoS One, vol.6, issue.4, p.19155

K. Higuchi, Genetic characterization of senescence-accelerated mouse (SAM), Exp Gerontol, vol.32, issue.1-2, pp.129-138, 1997.

G. Hilaire, Endogenous noradrenaline affects the maturation and function of the respiratory network: possible implication for SIDS, Auton Neurosci, vol.126, pp.320-331, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00300417

G. Hilaire and B. Duron, Maturation of the mammalian respiratory system, Physiol Rev, vol.79, issue.2, pp.325-360, 1999.
URL : https://hal.archives-ouvertes.fr/hal-00290938

G. Hilaire, N. Voituron, C. Menuet, R. M. Ichiyama, H. H. Subramanian et al.,

. Dutschmann, The role of serotonin in respiratory function and dysfunction, Respir Physiol Neurobiol, vol.174, issue.1-2, pp.76-88, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00625284

H. Hioki, F. Fujiyama, K. Nakamura, S. X. Wu, W. Matsuda et al., , 2004.

, Chemically specific circuit composed of vesicular glutamate transporter 3-and preprotachykinin B-producing interneurons in the rat neocortex, Cereb Cortex, vol.14, issue.11

H. Hioki, H. Nakamura, Y. F. Ma, M. Konno, T. Hayakawa et al., Vesicular glutamate transporter 3-expressing nonserotonergic projection neurons constitute a subregion in the rat midbrain raphe nuclei, J Comp Neurol, vol.518, issue.5, pp.668-686, 2010.

M. R. Hodges, S. Best, and G. B. Richerson, Altered ventilatory and thermoregulatory control in male and female adult Pet-1 null mice, Respir Physiol Neurobiol, vol.177, issue.2, pp.133-140, 2011.

N. Holler, R. Zaru, O. Micheau, M. Thome, A. Attinger et al.,

B. Schneider, J. Seed, and . Tschopp, Fas triggers an alternative, caspase-8independent cell death pathway using the kinase RIP as effector molecule, Nat Immunol, vol.1, issue.6, pp.489-495, 2000.

S. L. Hong, A. M. Estrada-sanchez, S. J. Barton, and G. V. Rebec, Early exposure to dynamic environments alters patterns of motor exploration throughout the lifespan, Behav Brain Res, vol.302, pp.81-87, 2016.

M. Hosokawa, T. Abe, K. Higuchi, K. Shimakawa, Y. Omori et al., Management and design of the maintenance of SAM mouse strains: an animal model for accelerated senescence and age-associated disorders, Exp Gerontol, vol.32, issue.1-2, pp.111-116, 1997.

M. Hosokawa, H. Fujisawa, B. H. Zhu, H. Jujo, and K. Higuchi, In vitro study of the mechanisms of senescence acceleration, Exp Gerontol, vol.32, issue.1-2, pp.197-203, 1997.

Y. T. Hsu, K. G. Wolter, and R. J. Youle, Cytosol-to-membrane redistribution of Bax and Bcl-X(L) during apoptosis, Proc Natl Acad Sci U S A, vol.94, issue.8, pp.3668-3672, 1997.

Z. Y. Hu, G. Liu, X. R. Cheng, Y. Huang, S. Yang et al.,

. Zhang, JD-30, an active fraction extracted from Danggui-Shaoyao-San, decreases beta-amyloid content and deposition, improves LTP reduction and prevents spatial cognition impairment in SAMP8 mice, Exp Gerontol, vol.47, issue.1, pp.14-22, 2012.

A. M. Hudson and L. Cooley, Drosophila Kelch functions with Cullin-3 to organize the ring canal actin cytoskeleton, J Cell Biol, vol.188, issue.1, pp.29-37, 2010.

C. Y. Huh, M. Danik, F. Manseau, L. E. Trudeau, and S. Williams, Chronic exposure to nerve growth factor increases acetylcholine and glutamate release from cholinergic neurons of the rat medial septum and diagonal band of Broca via mechanisms mediated by p75NTR, J Neurosci, vol.28, issue.6, pp.1404-1409, 2008.

Y. Ihara, Neurofibrillary tangles/paired helical filaments (1981-83), 2006.
DOI : 10.3233/jad-2006-9s324

, Alzheimers Dis, vol.9, pp.209-217

T. Ishikawa, Y. Sahara, and T. Takahashi, A single packet of transmitter does not saturate postsynaptic glutamate receptors, Neuron, vol.34, issue.4, pp.613-621, 2002.

Z. Jin, Y. Li, R. Pitti, D. Lawrence, V. C. Pham et al., , 2009.

, Cullin3-based polyubiquitination and p62-dependent aggregation of caspase-8 mediate extrinsic apoptosis signaling, Cell, vol.137, issue.4, pp.721-735

B. C. Jost and G. T. Grossberg, The evolution of psychiatric symptoms in, 1996.

, Alzheimer's disease: a natural history study, J Am Geriatr Soc, vol.44, issue.9, pp.1078-1081

P. J. Jost, S. Grabow, D. Gray, M. D. Mckenzie, U. Nachbur et al.,

E. Thomas, C. Borner, J. Silke, A. Strasser, and T. Kaufmann, XIAP discriminates between type I and type II FAS-induced apoptosis, Nature, vol.460, issue.7258, pp.1035-1039, 2009.

N. Juge, Y. Yoshida, S. Yatsushiro, H. Omote, and Y. Moriyama, Vesicular glutamate transporter contains two independent transport machineries, J Biol Chem, vol.281, issue.51, pp.39499-39506, 2006.

T. Kaneko and F. Fujiyama, Complementary distribution of vesicular glutamate transporters in the central nervous system, Neurosci Res, vol.42, issue.4, pp.243-250, 2002.

R. Kang, H. J. Zeh, M. T. Lotze, and D. Tang, The Beclin 1 network regulates autophagy and apoptosis, Cell Death Differ, vol.18, issue.4, pp.571-580, 2011.

Y. H. Kao, L. Lassova, T. Bar-yehuda, R. H. Edwards, P. Sterling et al., , 2004.

, Evidence that certain retinal bipolar cells use both glutamate and GABA, J Comp Neurol, vol.478, issue.3, pp.207-218

N. Karasawa, K. Nagatsu, T. Sakai, K. Nagatsu, M. Watanabe et al., , 1997.

, Immunocytochemical study of catecholaminergic neurons in the senescenceaccelerated mouse (SAM-P8) brain, J Neural Transm (Vienna), vol.104, pp.1267-1275

A. Kashani, C. Betancur, B. Giros, E. Hirsch, and S. E. Mestikawy, Altered expression of vesicular glutamate transporters VGLUT1 and VGLUT2 in Parkinson disease, Neurobiol Aging, vol.28, issue.4, pp.568-578, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00154872

A. Kashani, E. Lepicard, O. Poirel, C. Videau, J. P. David et al.,

B. Delacourte, J. Giros, C. Epelbaum, S. E. Betancur, and . Mestikawy, Loss of VGLUT1 and VGLUT2 in the prefrontal cortex is correlated with cognitive decline in Alzheimer disease, Neurobiol Aging, vol.29, issue.11, pp.1619-1630, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00166908

S. Kaushik, U. Bandyopadhyay, S. Sridhar, R. Kiffin, M. Martinez-vicente et al.,

E. Orenstein, A. M. Wong, and . Cuervo, Chaperone-mediated autophagy at a glance, J Cell Sci, vol.124, pp.495-499, 2011.

T. Kawamata, K. Akiguchi, C. Maeda, K. Tanaka, M. Higuchi et al.,

. Takeda, Age-related changes in the brains of senescence-accelerated mice (SAM): association with glial and endothelial reactions, Microsc Res Tech, vol.43, issue.1, pp.59-67, 1998.

J. F. Kerr, A. H. Wyllie, and A. R. Currie, Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics, Br J Cancer, vol.26, issue.4, p.239, 1972.

D. Khimich, R. Nouvian, R. Pujol, S. Tom-dieck, A. Egner et al.,

. Moser, Hair cell synaptic ribbons are essential for synchronous auditory signalling, Nature, vol.434, issue.7035, pp.889-894, 2005.

H. C. Kinney, G. B. Richerson, S. M. Dymecki, R. A. Darnall, and E. E. Nattie, , 2009.

, The brainstem and serotonin in the sudden infant death syndrome, Annu Rev Pathol, vol.4, pp.517-550

Y. Kiraz, A. Adan, M. Yandim, and Y. Baran, Major apoptotic mechanisms and genes involved in apoptosis, Tumour Biol, vol.37, issue.7, pp.8471-8486, 2016.
DOI : 10.1007/s13277-016-5035-9

Y. Kitamura, X. H. Zhao, T. Ohnuki, and Y. Nomura, Ligand-binding characteristics of, 1989.

Y. Kitamura, X. H. Zhao, T. Ohnuki, M. Takei, and Y. Nomura, Age-related changes in transmitter glutamate and NMDA receptor/channels in the brain of senescence-accelerated mouse, Neurosci Lett, vol.137, issue.2, pp.169-172, 1992.

M. Kolodziejczak, C. Bechade, N. Gervasi, T. Irinopoulou, S. M. Banas et al.,

A. Rebsam, L. Roumier, and . Maroteaux, Serotonin Modulates Developmental Microglia via 5-HT2B Receptors: Potential Implication during Synaptic Refinement of Retinogeniculate Projections, ACS Chem Neurosci, vol.6, issue.7, pp.1219-1230, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01224546

C. Kraft and S. Martens, Mechanisms and regulation of autophagosome formation, Curr Opin Cell Biol, vol.24, issue.4, pp.496-501, 2012.

T. Kraus, W. L. Neuhuber, and M. Raab, , 2004.

, immunoreactivity in motor endplates of striated esophageal but not skeletal muscles in the mouse, Neurosci Lett, vol.360, issue.1-2, pp.53-56

T. Kuwana, M. R. Mackey, G. Perkins, M. H. Ellisman, M. Latterich et al.,

D. D. Green and . Newmeyer, Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane, Cell, vol.111, issue.3, pp.331-342, 2002.

S. I. Kwon, H. J. Cho, J. H. Jung, K. Yoshimoto, K. Shirasu et al., , 2010.

, Rab GTPase RabG3b functions in autophagy and contributes to tracheary element differentiation in Arabidopsis, Plant J, vol.64, issue.1, pp.151-164

E. Laane, K. P. Tamm, E. Buentke, K. Ito, P. Kharaziha et al.,

K. Bjorklund, J. Hultenby, M. Lundin, S. Heyman, J. Soderhall et al.,

B. Pandolfi, T. Zhivotovsky, D. Panaretakis, and . Grander, Cell death induced by dexamethasone in lymphoid leukemia is mediated through initiation of autophagy, Cell Death Differ, vol.16, issue.7, pp.1018-1029, 2009.

P. N. Lacor, M. C. Buniel, L. Chang, S. J. Fernandez, Y. Gong et al., Synaptic targeting by Alzheimer's-related amyloid beta oligomers, J Neurosci, vol.24, issue.45, p.10191, 2004.

Y. Lee, H. R. Kim, and S. C. Ahn, Vesicular glutamate transporter 3 is strongly upregulated in cochlear inner hair cells and spiral ganglion cells of developing circling mice, Neurosci Lett, vol.584, pp.320-324, 2015.

Y. R. Lee, W. C. Yuan, H. C. Ho, C. H. Chen, H. M. Shih et al., The Cullin 3 substrate adaptor KLHL20 mediates DAPK ubiquitination to control interferon responses, EMBO J, vol.29, issue.10, pp.1748-1761, 2010.

C. Leranth, M. Shanabrough, and T. L. Horvath, Hormonal regulation of hippocampal spine synapse density involves subcortical mediation, Neuroscience, vol.101, issue.2, pp.349-356, 2000.

S. Levay, M. P. Stryker, and C. J. Shatz, Ocular dominance columns and their development in layer IV of the cat's visual cortex: a quantitative study, J Comp Neurol, vol.179, issue.1, pp.223-244, 1978.

B. Levine, S. Sinha, and G. Kroemer, Bcl-2 family members: dual regulators of apoptosis and autophagy, Autophagy, vol.4, issue.5, pp.600-606, 2008.

G. Li, H. Cheng, X. Zhang, X. Shang, H. Xie et al., , 2013.

, Hippocampal neuron loss is correlated with cognitive deficits in SAMP8 mice, Neurol Sci, vol.34, issue.6, pp.963-969

W. W. Li, J. Li, and J. K. Bao, Microautophagy: lesser-known self-eating, Cell Mol Life Sci, vol.69, issue.7, pp.1125-1136, 2012.

M. C. Liberman, Single-neuron labeling in the cat auditory nerve, Science, vol.216, issue.4551, pp.1239-1241, 1982.

M. C. Liberman, L. W. Dodds, and S. Pierce, Afferent and efferent innervation of the cat cochlea: quantitative analysis with light and electron microscopy, J Comp Neurol, vol.301, issue.3, pp.443-460, 1990.

H. W. Lin, A. C. Furman, S. G. Kujawa, and M. C. Liberman, Primary neural degeneration in the Guinea pig cochlea after reversible noise-induced threshold shift, J Assoc Res Otolaryngol, vol.12, issue.5, pp.605-616, 2011.

E. Littré, Le Nouveau petit Littré, 2009.

G. Liu, Z. K. Wang, Z. Y. Wang, D. B. Yang, Z. P. Liu et al., , 2016.

S. Y. Liu, C. L. Chen, T. T. Yang, W. C. Huang, C. Y. Hsieh et al., Albumin prevents reactive oxygen species-induced mitochondrial damage, autophagy, and apoptosis during serum starvation, Apoptosis, vol.17, issue.11, pp.1156-1169, 2012.

X. A. Liu, J. Song, Q. Jiang, Q. Wang, Q. Tian et al., Expression of the hyperphosphorylated tau attenuates ER stress-induced apoptosis with upregulation of unfolded protein response, Apoptosis, vol.17, issue.10, pp.1039-1049, 2012.

J. C. Lopez-ramos, M. T. Jurado-parras, C. Sanfeliu, D. Acuna-castroviejo, and J. ,

. Delgado-garcia, Learning capabilities and CA1-prefrontal synaptic plasticity in a mice model of accelerated senescence, Neurobiol Aging, vol.33, issue.3, pp.627-613, 2012.

M. F. Luciani, C. Giusti, B. Harms, Y. Oshima, H. Kikuchi et al., Atg1 allows second-signaled autophagic cell death in Dictyostelium, Autophagy, vol.7, issue.5, pp.501-508, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00608345

S. Luo, M. Garcia-arencibia, R. Zhao, C. Puri, P. P. Toh et al., Bim inhibits autophagy by recruiting Beclin 1 to microtubules, Mol Cell, vol.47, issue.3, pp.359-370, 2012.

S. Luo and D. C. Rubinsztein, Apoptosis blocks Beclin 1-dependent autophagosome synthesis: an effect rescued by Bcl-xL, Cell Death Differ, vol.17, issue.2, p.268, 2010.

X. Luo, H. Budihardjo, C. Zou, X. Slaughter, and . Wang, Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors, Cell, vol.94, issue.4, pp.481-490, 1998.

S. A. Lyness, C. Zarow, and H. C. Chui, Neuron loss in key cholinergic and aminergic nuclei in Alzheimer disease: a meta-analysis, Neurobiol Aging, vol.24, issue.1, pp.1-23, 2003.

K. Mahati, V. Bhagya, T. Christofer, A. Sneha, and B. S. Shankaranarayana-rao, , 2016.

, Enriched environment ameliorates depression-induced cognitive deficits and restores abnormal hippocampal synaptic plasticity, Neurobiol Learn Mem, vol.134, pp.379-391

M. Mainardi, A. D. Garbo, M. Caleo, N. Berardi, A. Sale et al., , 2014.

, Environmental enrichment strengthens corticocortical interactions and reduces amyloid-beta oligomers in aged mice, Front Aging Neurosci, vol.6, p.1

D. M. Mann, P. O. Yates, and B. Marcyniuk, Correlation between senile plaque and neurofibrillary tangle counts in cerebral cortex and neuronal counts in cortex and subcortical structures in Alzheimer's disease, Neurosci Lett, vol.56, issue.1, pp.51-55, 1985.

A. L. Markowska, E. L. Spangler, and D. K. Ingram, Behavioral assessment of the senescence-accelerated mouse (SAM P8 and R1), Physiol Behav, vol.64, issue.1, pp.15-26, 1998.

R. T. Marquez and L. Xu, Bcl-2:Beclin 1 complex: multiple, mechanisms regulating autophagy/apoptosis toggle switch, Am J Cancer Res, vol.2, issue.2, pp.214-221, 2012.

A. Martini, M. Mazzoli, M. Rosignoli, P. Trevisi, S. Maggi et al., Hearing in the elderly: a population study, Audiology, vol.40, issue.6, pp.285-293, 2001.

E. Masliah and R. Terry, The role of synaptic proteins in the pathogenesis of disorders of the central nervous system, Brain Pathol, vol.3, issue.1, pp.77-85, 1993.

A. Massie, A. Schallier, K. Vermoesen, L. Arckens, and Y. Michotte, Biphasic and bilateral changes in striatal VGLUT1 and 2 protein expression in hemi-Parkinson rats, Neurochem Int, vol.57, issue.2, pp.111-118, 2010.

M. Matjusaitis, G. Chin, E. A. Sarnoski, and A. Stolzing, Biomarkers to identify and isolate senescent cells, Ageing Res Rev, vol.29, pp.1-12, 2016.

T. J. Matthews and M. F. Macdorman, Infant mortality statistics from the 2010 period linked birth/infant death data set, Natl Vital Stat Rep, vol.62, issue.8, pp.1-26, 2013.

P. R. Maycox, T. Deckwerth, J. W. Hell, and R. Jahn, Glutamate uptake by brain synaptic vesicles. Energy dependence of transport and functional reconstitution in proteoliposomes, J Biol Chem, vol.263, issue.30, pp.15423-15428, 1988.

C. K. Mcphee and E. H. Baehrecke, The engulfment receptor Draper is required for autophagy during cell death, Autophagy, vol.6, issue.8, pp.1192-1193, 2010.

H. C. Meeker, K. K. Chadman, A. T. Heaney, and R. I. Carp, Assessment of social interaction and anxiety-like behavior in senescence-accelerated-prone andresistant mice, Physiol Behav, vol.118, pp.97-102, 2013.

S. Melchionda, N. Ahituv, L. Bisceglia, T. Sobe, F. Glaser et al.,

. Gasparini, MYO6, the human homologue of the gene responsible for deafness in, 2001.

, Snell's waltzer mice, is mutated in autosomal dominant nonsyndromic hearing loss, Am J Hum Genet, vol.69, issue.3, pp.635-640

J. Menardo, Y. Tang, S. Ladrech, M. Lenoir, F. Casas et al.,

T. Rebillard, J. L. Maurice, J. Puel, and . Wang, Oxidative stress, inflammation, and autophagic stress as the key mechanisms of premature age-related hearing loss in SAMP8 mouse Cochlea, Antioxid Redox Signal, vol.16, issue.3, pp.263-274, 2012.

C. Menuet, N. Kourdougli, G. Hilaire, and N. Voituron, Differences in serotoninergic metabolism possibly contribute to differences in breathing phenotype of FVB/N and C57BL/6J mice, J Appl Physiol, vol.110, issue.6, pp.1572-1581, 1985.

A. C. Meyer, T. Frank, D. Khimich, G. Hoch, D. Riedel et al., Tuning of synapse number, structure and function in the cochlea, Nat Neurosci, vol.12, issue.4, pp.444-453, 2009.

H. Mistry, B. A. Wilson, I. J. Roberts, C. J. O'kane, and J. B. Skeath, Cullin-3 regulates pattern formation, external sensory organ development and cell survival during Drosophila development, Mech Dev, vol.121, issue.12, pp.1495-1507, 2004.

A. B. Mitnitski, A. J. Mogilner, and K. Rockwood, Accumulation of deficits as a proxy measure of aging, ScientificWorldJournal, vol.1, pp.323-336, 2001.

T. Miyaji, N. Echigo, M. Hiasa, S. Senoh, H. Omote et al., , 2008.

, Identification of a vesicular aspartate transporter, Proc Natl Acad Sci U S A, vol.105, issue.33

M. Miyamoto, Characteristics of age-related behavioral changes in senescence-accelerated mouse SAMP8 and SAMP10, Exp Gerontol, vol.32, issue.1-2, pp.139-148, 1997.

M. Miyamoto, Y. Kiyota, M. Nishiyama, and A. Nagaoka, Senescenceaccelerated mouse (SAM): age-related reduced anxiety-like behavior in the SAM-P, 1992.

, Physiol Behav, vol.51, issue.5, pp.979-985

M. Miyamoto, Y. Kiyota, N. Yamazaki, A. Nagaoka, T. Matsuo et al.,

. Takeda, Age-related changes in learning and memory in the senescenceaccelerated mouse (SAM), Physiol Behav, vol.38, issue.3, pp.399-406, 1986.

D. Moechars, M. C. Weston, S. Leo, Z. Callaerts-vegh, I. Goris et al.,

P. Cik, S. Van-der-spek, T. Kass, R. Meert, C. D'hooge et al., Vesicular glutamate transporter VGLUT2 expression levels control quantal size and neuropathic pain, J Neurosci, vol.26, pp.12055-12066, 2006.

F. Mora, G. Segovia, and A. Del-arco, Aging, plasticity and environmental enrichment: structural changes and neurotransmitter dynamics in several areas of the brain, Brain Res Rev, vol.55, issue.1, pp.78-88, 2007.

A. Mora-gallegos, M. Rojas-carvajal, S. Salas, A. Saborio-arce, J. Fornaguera-trias et al., Age-dependent effects of environmental enrichment on spatial memory and neurochemistry, Neurobiol Learn Mem, vol.118, pp.96-104, 2015.

J. E. Morley, The SAMP8 mouse: a model of Alzheimer disease?, Biogerontology, vol.3, issue.1-2, pp.57-60, 2002.

J. E. Morley, V. B. Kumar, A. E. Bernardo, S. A. Farr, K. Uezu et al., Beta-amyloid precursor polypeptide in SAMP8 mice affects learning and memory, Peptides, vol.21, issue.12, pp.1761-1767, 2000.

S. F. Morrison, K. Nakamura, and C. J. Madden, Central control of thermogenesis in mammals, Exp Physiol, vol.93, issue.7, pp.773-797, 2008.

T. Moser and D. Beutner, Kinetics of exocytosis and endocytosis at the cochlear inner hair cell afferent synapse of the mouse, Proc Natl Acad Sci U S A, vol.97, issue.2, pp.883-888, 2000.

S. Mukhopadhyay, P. K. Panda, N. Sinha, D. N. Das, and S. K. Bhutia, , 2014.

, Autophagy and apoptosis: where do they meet?, Apoptosis, vol.19, issue.4, pp.555-566

S. Naito and T. Ueda, Characterization of glutamate uptake into synaptic vesicles, J Neurochem, vol.44, issue.1, pp.99-109, 1985.

H. Nakahara, T. Kanno, Y. Inai, K. Utsumi, M. Hiramatsu et al., Mitochondrial dysfunction in the senescence accelerated mouse (SAM), Free Radic Biol Med, vol.24, issue.1, pp.85-92, 1998.

K. Nakamura, H. Hioki, F. Fujiyama, and T. Kaneko, Postnatal changes of vesicular glutamate transporter (VGluT)1 and VGluT2 immunoreactivities and their colocalization in the mouse forebrain, J Comp Neurol, vol.492, issue.3, pp.263-288, 2005.

K. Nakamura, S. X. Wu, F. Fujiyama, K. Okamoto, H. Hioki et al., , 2004.

, Independent inputs by VGLUT2-and VGLUT3-positive glutamatergic terminals onto rat sympathetic preganglionic neurons, Neuroreport, vol.15, issue.3, pp.431-436

G. Nardai, P. Csermely, and C. Soti, Chaperone function and chaperone overload in the aged. A preliminary analysis, Exp Gerontol, vol.37, pp.1257-1262, 2002.

A. R. Nelson, M. D. Sweeney, A. P. Sagare, and B. V. Zlokovic, Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer's disease, Biochim Biophys Acta, vol.1862, issue.5, pp.887-900, 2016.

N. R. Nemzou, A. V. Bulankina, D. Khimich, A. Giese, and T. Moser, Synaptic organization in cochlear inner hair cells deficient for the CaV1.3 (alpha1D) subunit of Ltype Ca2+ channels, Neuroscience, vol.141, issue.4, pp.1849-1860, 2006.

T. P. Neufeld, TOR-dependent control of autophagy: biting the hand that feeds, Curr Opin Cell Biol, vol.22, issue.2, pp.157-168, 2010.

B. Ni, P. R. Rosteck, N. S. Nadi, and S. M. Paul, Cloning and expression of a cDNA encoding a brain-specific Na(+)-dependent inorganic phosphate cotransporter, Proc Natl Acad Sci U S A, vol.91, issue.12, pp.5607-5611, 1994.

A. Nickerson-poulin, A. Guerci, S. E. Mestikawy, and K. Semba, Vesicular glutamate transporter 3 immunoreactivity is present in cholinergic basal forebrain neurons projecting to the basolateral amygdala in rat, J Comp Neurol, vol.498, issue.5, pp.690-711, 2006.

T. Nishi, N. Shimizu, M. Hiramoto, I. Sato, Y. Yamaguchi et al.,

K. Tanaka, H. Kataoka, H. Watanabe, and . Handa, Spatial redox regulation of a critical cysteine residue of NF-kappa B in vivo, J Biol Chem, vol.277, pp.44548-44556, 2002.

Y. Nishida, S. Arakawa, K. Fujitani, H. Yamaguchi, T. Mizuta et al.,

K. Otsu, Y. Tsujimoto, and S. Shimizu, Discovery of Atg5/Atg7-independent alternative macroautophagy, Nature, vol.461, issue.7264, pp.654-658, 2009.

H. Nishimaru, C. E. Restrepo, J. Ryge, Y. Yanagawa, and O. Kiehn, Mammalian motor neurons corelease glutamate and acetylcholine at central synapses, Proc Natl Acad Sci U S A, vol.102, issue.14, pp.5245-5249, 2005.

K. Nishizawa, H. Saito, and N. Nishiyama, Effects of Kamikihi-To, a traditional Chinese medicine, on passive and conditioned avoidance performance impairment in senescence accelerated mouse (SAM), Neurobiol Learn Mem, vol.54, issue.4, pp.200-210, 1990.

D. Njus, P. M. Kelley, and G. J. Harnadek, Bioenergetics of secretory vesicles, 1986.

, Biochim Biophys Acta, vol.853, issue.3-4, pp.237-265

Y. Nomura, Y. Kitamura, T. Ohnuki, T. Arima, Y. Yamanaka et al., Alterations in acetylcholine, NMDA, benzodiazepine receptors and protein kinase C in the brain of the senescence-accelerated mouse: an animal model useful for studies on cognitive enhancers, Behav Brain Res, vol.83, issue.1-2, pp.51-55, 1997.

Y. Nomura and Y. Okuma, Age-related defects in lifespan and learning ability in SAMP8 mice, Neurobiol Aging, vol.20, issue.2, pp.111-115, 1999.

R. Nouvian, Physiologie de l'audition, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00094975

R. Nouvian, D. Beutner, T. D. Parsons, and T. Moser, Structure and function of the hair cell ribbon synapse, J Membr Biol, vol.209, issue.2-3, pp.153-165, 2006.

R. Nouvian, J. Neef, A. V. Bulankina, E. Reisinger, T. Pangrsic et al.,

T. Brose, T. Binz, and . Moser, Exocytosis at the hair cell ribbon synapse apparently operates without neuronal SNARE proteins, Nat Neurosci, vol.14, issue.4, pp.411-413, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00619269

A. Oberstein, P. D. Jeffrey, and Y. Shi, Crystal structure of the Bcl-XL-Beclin, vol.1, 2007.

, Beclin 1 is a novel BH3-only protein, J Biol Chem, vol.282, issue.17, p.13123

K. Ogita, K. Hirata, D. G. Bole, S. Yoshida, Y. Tamura et al., Inhibition of vesicular glutamate storage and exocytotic release by Rose Bengal, J Neurochem, vol.77, issue.1, pp.34-42, 2001.

A. Ohta, N. Akiguchi, K. Seriu, H. Ohnishi, K. Yagi et al., Deterioration in learning and memory of fear conditioning in response to context in aged SAMP8 mice, Neurobiol Aging, vol.22, issue.3, pp.479-484, 2001.

A. L. Oliveira, F. Hydling, E. Olsson, T. Shi, R. H. Edwards et al.,

S. Hokfelt, B. Cullheim, and . Meister, Cellular localization of three vesicular glutamate transporter mRNAs and proteins in rat spinal cord and dorsal root ganglia, Synapse, vol.50, issue.2, pp.117-129, 2003.

H. Omote, T. Miyaji, N. Juge, and Y. Moriyama, Vesicular neurotransmitter transporter: bioenergetics and regulation of glutamate transport, Biochemistry, vol.50, 2011.

H. Onimaru and I. Homma, A novel functional neuron group for respiratory rhythm generation in the ventral medulla, J Neurosci, vol.23, issue.4, pp.1478-1486, 2003.

K. Op-de-beeck, J. Schacht, and G. Van-camp, Apoptosis in acquired and genetic hearing impairment: the programmed death of the hair cell, Hear Res, vol.281, issue.1-2, pp.18-27, 2011.

P. L. Opresko and J. W. Shay, Telomere-associated aging disorders, Ageing Res Rev, 2016.

T. Osaka, Hypoxia-induced hypothermia mediated by noradrenaline and nitric oxide in the rostromedial preoptic area, Neuroscience, vol.179, pp.170-178, 2011.

E. N. Ottem, J. G. Godwin, S. Krishnan, and S. L. Petersen, Dual-phenotype, 2004.

, GABA/glutamate neurons in adult preoptic area: sexual dimorphism and function, J Neurosci, vol.24, issue.37, pp.8097-8105

L. Ouyang, Z. Shi, S. Zhao, F. T. Wang, T. T. Zhou et al., , 2012.

, Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis, Cell Prolif, vol.45, issue.6, pp.487-498

M. Pallas, A. Camins, M. A. Smith, G. Perry, H. G. Lee et al., From aging to Alzheimer's disease: unveiling "the switch" with the senescence-accelerated mouse model (SAMP8), J Alzheimers Dis, vol.15, issue.4, pp.615-624, 2008.

M. Panchal, M. Rholam, and N. Brakch, Abnormalities of peptide metabolism in Alzheimer disease, Curr Neurovasc Res, vol.1, issue.4, pp.317-323, 2004.

T. Pangrsic, L. Lasarow, K. Reuter, H. Takago, M. Schwander et al.,

J. S. Tarantino, N. Bailey, N. Strenzke, U. Brose, E. Muller et al., , 2010.

, Hearing requires otoferlin-dependent efficient replenishment of synaptic vesicles in hair cells, Nat Neurosci, vol.13, issue.7, pp.869-876

S. M. Parsons, Transport mechanisms in acetylcholine and monoamine storage, FASEB J, vol.14, issue.15, pp.2423-2434, 2000.

D. S. Paterson, F. L. Trachtenberg, E. G. Thompson, R. A. Belliveau, A. H. Beggs et al.,

A. E. Darnall, H. F. Chadwick, H. C. Krous, and . Kinney, Multiple serotonergic brainstem abnormalities in sudden infant death syndrome, JAMA, vol.296, issue.17, pp.2124-2132, 2006.

S. Pattingre, L. Espert, M. Biard-piechaczyk, and P. Codogno, Regulation of macroautophagy by mTOR and Beclin 1 complexes, Biochimie, vol.90, issue.2, pp.313-323, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00257274

C. Pelegri, A. M. Canudas, J. Valle, G. Casadesus, M. A. Smith et al., Increased permeability of blood-brain barrier on the hippocampus of a murine model of senescence, Mech Ageing Dev, vol.128, issue.9, pp.522-528, 2007.

Y. Pena, M. Prunell, V. Dimitsantos, R. Nadal, and R. M. Escorihuela, , 2006.

, Behav Brain Res, vol.174, issue.1, pp.181-187

B. G. Peng, Q. X. Li, T. Y. Ren, S. Ahmad, S. P. Chen et al., Group I metabotropic glutamate receptors in spiral ganglion neurons contribute to excitatory neurotransmissions in the cochlea, Neuroscience, vol.123, issue.1, pp.221-230, 2004.

Z. Peng, G. P. Wang, R. Zeng, J. Y. Guo, C. F. Chen et al., , 2013.

, Temporospatial expression and cellular localization of VGLUT3 in the rat cochlea, Brain Res, vol.1537, pp.100-110

L. M. Peters, D. W. Anderson, A. J. Griffith, K. M. Grundfast, and T. B. ,

T. B. Madeo, R. J. Friedman, and . Morell, Mutation of a transcription factor, TFCP2L3, causes progressive autosomal dominant hearing loss, DFNA28, Hum Mol Genet, vol.11, issue.23, pp.2877-2885, 2002.

N. Pietrancosta, A. Kessler, F. C. Favre-besse, N. Triballeau, T. Quentin et al., Rose Bengal analogs and vesicular glutamate transporters (VGLUTs), Bioorg Med Chem, vol.18, issue.18, pp.6922-6933, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02351315

J. L. Puel, Chemical synaptic transmission in the cochlea, Prog Neurobiol, vol.47, issue.6, pp.449-476, 1995.

J. L. Puel, R. Pujol, F. Tribillac, S. Ladrech, and M. Eybalin, Excitatory amino acid antagonists protect cochlear auditory neurons from excitotoxicity, J Comp Neurol, vol.341, issue.2, pp.241-256, 1994.

L. Radoshevich, L. Murrow, N. Chen, E. Fernandez, S. Roy et al., ATG12 conjugation to ATG3 regulates mitochondrial homeostasis and cell death, Cell, vol.142, issue.4, pp.590-600, 2010.

S. Ramirez, T. Allen, L. Villagracia, Y. Chae, J. M. Ramirez et al., , 2016.

, Inner ear lesion and the differential roles of hypoxia and hypercarbia in triggering active movements: Potential implication for the Sudden Infant Death Syndrome, Neuroscience, vol.337, pp.9-16

G. L. Reijnierse, H. Veldstra, C. J. Van-den, and . Berg, Subcellular localization of gamma-aminobutyrate transaminase and glutamate dehydrogenase in adult rat brain. Evidence for at least two small glutamate compartments in brain, Biochem J, vol.152, issue.3, pp.469-475, 1975.

J. Ren, C. Qin, F. Hu, J. Tan, L. Qiu et al., Habenula "cholinergic" neurons co-release glutamate and acetylcholine and activate postsynaptic neurons via distinct transmission modes, Neuron, vol.69, issue.3, pp.445-452, 2011.

T. T. Renault, O. Teijido, B. Antonsson, L. M. Dejean, and S. Manon, , 2013.

, Regulation of Bax mitochondrial localization by Bcl-2 and Bcl-x(L): keep your friends close but your enemies closer, Int J Biochem Cell Biol, vol.45, issue.1, pp.64-67

G. Riedel and K. G. Reymann, Metabotropic glutamate receptors in hippocampal long-term potentiation and learning and memory, Acta Physiol Scand, vol.157, issue.1, pp.1-19, 1996.

C. M. Robinson, J. Y. Jung, and G. J. Nau, Interferon-gamma, tumor necrosis factor, and interleukin-18 cooperate to control growth of Mycobacterium tuberculosis in human macrophages, Cytokine, vol.60, issue.1, pp.233-241, 2012.

K. Rockwood and A. Mitnitski, Frailty in relation to the accumulation of deficits, J Gerontol A Biol Sci Med Sci, vol.62, issue.7, pp.722-727, 2007.

R. Romero-garcia, M. Atienza, and J. L. Cantero, Predictors of coupling between structural and functional cortical networks in normal aging, Hum Brain Mapp, vol.35, issue.6, pp.2724-2740, 2014.

S. Roseth, E. M. Fykse, and F. Fonnum, Uptake of L-glutamate into rat brain synaptic vesicles: effect of inhibitors that bind specifically to the glutamate transporter, J Neurochem, vol.65, issue.1, pp.96-103, 1995.

D. L. Rosin, D. A. Chang, and P. G. Guyenet, Afferent and efferent connections of the rat retrotrapezoid nucleus, J Comp Neurol, vol.499, issue.1, pp.64-89, 2006.
DOI : 10.1002/cne.21105

A. D. Rubinstein, M. Eisenstein, Y. Ber, S. Bialik, and A. Kimchi, The autophagy protein Atg12 associates with antiapoptotic Bcl-2 family members to promote mitochondrial apoptosis, Mol Cell, vol.44, issue.5, pp.698-709, 2011.
DOI : 10.1016/j.molcel.2011.10.014

URL : https://doi.org/10.1016/j.molcel.2011.10.014

J. Ruel, R. P. Bobbin, D. Vidal, R. Pujol, and J. L. Puel, The selective AMPA receptor antagonist GYKI 53784 blocks action potential generation and excitotoxicity in the guinea pig cochlea, Neuropharmacology, vol.39, issue.11, pp.1959-1973, 2000.
DOI : 10.1016/s0028-3908(00)00069-1

URL : https://hal.archives-ouvertes.fr/inserm-00267756

J. Ruel, S. Emery, R. Nouvian, T. Bersot, B. Amilhon et al.,

M. Lenoir, M. Eybalin, B. Delprat, T. A. Sivakumaran, B. Giros et al., Impairment of SLC17A8 encoding vesicular glutamate transporter-3, VGLUT3, underlies nonsyndromic deafness DFNA25 and inner hair cell dysfunction in null mice, Am J Hum Genet, vol.83, issue.2, pp.278-292, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00349467

J. Ruel, R. Nouvian, C. Gervais-d'aldin, R. Pujol, M. Eybalin et al., , 2001.

, Dopamine inhibition of auditory nerve activity in the adult mammalian cochlea, Eur J Neurosci, vol.14, issue.6, pp.977-986

J. Ruel, J. Wang, D. Dememes, S. Gobaille, J. L. Puel et al., , 2006.

, Dopamine transporter is essential for the maintenance of spontaneous activity of auditory nerve neurones and their responsiveness to sound stimulation, J Neurochem, vol.97, issue.1, pp.190-200

S. Safieddine and M. Eybalin, Expression of mGluR1 alpha mRNA receptor in rat and guinea pig cochlear neurons, Neuroreport, vol.7, issue.1, pp.193-196, 1995.

C. Sagne, S. E. Mestikawy, M. F. Isambert, M. Hamon, J. P. Henry et al.,

. Gasnier, Cloning of a functional vesicular GABA and glycine transporter by screening of genome databases, FEBS Lett, vol.417, issue.2, pp.177-183, 1997.

D. Y. Sakae, F. Marti, S. Lecca, F. Vorspan, E. Martin-garcia et al.,

A. Gutierrez-cuesta, N. Besnard, E. Heck, S. Herzog, V. F. Bolte et al.,

C. B. Bellivier, S. Eap, P. Crettol, J. Vanhoutte, A. Caboche et al.,

S. Maldonado, M. Daumas, S. Mameli, S. E. Jamain, and . Mestikawy, The absence of VGLUT3 predisposes to cocaine abuse by increasing dopamine and glutamate signaling in the nucleus accumbens, Mol Psychiatry, vol.20, issue.11, pp.1448-1459, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01536677

A. Salminen and K. Kaarniranta, Regulation of the aging process by autophagy, Trends Mol Med, vol.15, issue.5, pp.217-224, 2009.

W. Samuel, R. D. Terry, R. Deteresa, N. Butters, and E. Masliah, Clinical correlates of cortical and nucleus basalis pathology in Alzheimer dementia, Arch Neurol, vol.51, issue.8, pp.772-778, 1994.

E. Sato, T. Kurokawa, N. Oda, and S. Ishibashi, , 1996.

E. Sato, N. Oda, N. Ozaki, S. Hashimoto, T. Kurokawa et al., Early and transient increase in oxidative stress in the cerebral cortex of senescenceaccelerated mouse, Mech Ageing Dev, vol.86, issue.2, pp.105-114, 1996.

M. K. Schafer, H. Varoqui, N. Defamie, E. Weihe, and J. D. Erickson, Molecular cloning and functional identification of mouse vesicular glutamate transporter 3 and its expression in subsets of novel excitatory neurons, J Biol Chem, vol.277, issue.52, pp.50734-50748, 2002.

S. Schenck, S. M. Wojcik, N. Brose, and S. Takamori, A chloride conductance in VGLUT1 underlies maximal glutamate loading into synaptic vesicles, Nat Neurosci, vol.12, issue.2, pp.156-162, 2009.

F. Schmitz, A. Konigstorfer, and T. C. Sudhof, RIBEYE, a component of synaptic ribbons: a protein's journey through evolution provides insight into synaptic ribbon function, Neuron, vol.28, issue.3, pp.857-872, 2000.

R. P. Seal, O. Akil, E. Yi, C. M. Weber, L. Grant et al.,

E. Noebels, L. R. Glowatzki, R. H. Lustig, and . Edwards, Sensorineural deafness and seizures in mice lacking vesicular glutamate transporter 3, Neuron, vol.57, issue.2, pp.263-275, 2008.

G. Segovia, A. G. Yague, J. M. Garcia-verdugo, and F. Mora, Environmental enrichment promotes neurogenesis and changes the extracellular concentrations of glutamate and GABA in the hippocampus of aged rats, Brain Res Bull, vol.70, issue.1, pp.8-14, 2006.

A. Shamas-din, H. Brahmbhatt, B. Leber, and D. W. Andrews, BH3-only proteins: Orchestrators of apoptosis, Biochim Biophys Acta, vol.1813, issue.4, pp.508-520, 2011.

N. E. Sharpless and C. J. Sherr, Forging a signature of in vivo senescence, 2015.

, Nat Rev Cancer, vol.15, issue.7, pp.397-408

L. Sheets, J. G. Trapani, W. Mo, N. Obholzer, and T. Nicolson, Ribeye is required for presynaptic Ca(V)1.3a channel localization and afferent innervation of sensory hair cells, Development, vol.138, issue.7, pp.1309-1319, 2011.

H. M. Shen and P. Codogno, Autophagic cell death: Loch Ness monster or endangered species?, Autophagy, vol.7, issue.5, pp.457-465, 2011.

Y. C. Shen, D. L. Liao, C. L. Lu, J. Y. Chen, Y. J. Liou et al., , 2010.

, Resequencing of the vesicular glutamate transporter 2 gene (VGLUT2) reveals some rare genetic variants that may increase the genetic burden in schizophrenia, Schizophr Res, vol.121, issue.1-3, pp.179-186

S. Shimizu, T. Kanaseki, N. Mizushima, T. Mizuta, S. Arakawa-kobayashi et al.,

Y. Thompson and . Tsujimoto, Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes, Nat Cell Biol, vol.6, issue.12, pp.1221-1228, 2004.

F. Shutoh, A. Ina, S. Yoshida, J. Konno, and S. Hisano, Two distinct subtypes of serotonergic fibers classified by co-expression with vesicular glutamate transporter 3 in rat forebrain, Neurosci Lett, vol.432, issue.2, pp.132-136, 2008.

J. C. Smith, H. H. Ellenberger, K. Ballanyi, D. W. Richter, and J. L. Feldman, , 1991.

, Pre-Botzinger complex: a brainstem region that may generate respiratory rhythm in mammals, Science, vol.254, issue.5032, pp.726-729

J. Somogyi, A. Baude, Y. Omori, H. Shimizu, S. E. Mestikawy et al.,

M. Watanabe and P. Somogyi, GABAergic basket cells expressing cholecystokinin contain vesicular glutamate transporter type 3 (VGLUT3) in their synaptic terminals in hippocampus and isocortex of the rat, Eur J Neurosci, vol.19, issue.3, p.552, 2004.

S. L. Stella, S. Jr, A. Li, A. Sabatini, N. C. Vila et al., Comparison of the ontogeny of the vesicular glutamate transporter 3 (VGLUT3) with VGLUT1 and VGLUT2 in the rat retina, Brain Res, vol.1215, pp.20-29, 2008.

M. J. Stensrud, C. J. Sogn, and V. Gundersen, Immunogold characteristics of VGLUT3-positive GABAergic nerve terminals suggest corelease of glutamate, J Comp Neurol, vol.523, issue.18, pp.2698-2713, 2015.

R. L. Stornetta, C. P. Sevigny, and P. G. Guyenet, Vesicular glutamate transporter DNPI/VGLUT2 mRNA is present in C1 and several other groups of brainstem catecholaminergic neurons, J Comp Neurol, vol.444, issue.3, pp.191-206, 2002.

F. Strappazzon, M. Vietri-rudan, S. Campello, F. Nazio, F. Florenzano et al.,

B. Piacentini, F. Levine, and . Cecconi, Mitochondrial BCL-2 inhibits AMBRA1induced autophagy, EMBO J, vol.30, issue.7, pp.1195-1208, 2011.

P. Strata and R. Harvey, Dale's principle, Brain Res Bull, vol.50, issue.5-6, pp.349-350, 1999.

G. D. Stuber, T. S. Hnasko, J. P. Britt, R. H. Edwards, and A. Bonci, Neurosci, vol.30, issue.24, pp.8229-8233, 2010.

H. Sugiyama, H. Akiyama, I. Akiguchi, M. Kameyama, and T. Takeda, Loss of dendritic spines in hippocampal CA1 pyramidal cells in senescence accelerated mouse (SAM)-a quantitative Golgi study, Rinsho Shinkeigaku, vol.27, issue.7, pp.841-845, 1987.

D. Sulzer, M. P. Joyce, L. Lin, D. Geldwert, S. N. Haber et al., Dopamine neurons make glutamatergic synapses in vitro, J Neurosci, vol.18, 1998.

L. Sun, H. Wang, Z. Wang, S. He, S. Chen et al.,

. Wang, Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase, Cell, vol.148, issue.1-2, pp.213-227, 2012.

Q. J. Sun, R. G. Berkowitz, A. K. Goodchild, and P. M. Pilowsky, Serotonin inputs to inspiratory laryngeal motoneurons in the rat, J Comp Neurol, vol.451, issue.1, pp.91-98, 2002.

F. X. Sureda, J. Gutierrez-cuesta, M. Romeu, M. Mulero, A. M. Canudas et al.,

M. Mallol and . Pallas, Changes in oxidative stress parameters and neurodegeneration markers in the brain of the senescence-accelerated mice SAMP-8, Exp Gerontol, vol.41, issue.4, pp.360-367, 2006.

R. H. Swerdlow and S. M. Khan, A "mitochondrial cascade hypothesis" for sporadic Alzheimer's disease, Med Hypotheses, vol.63, issue.1, pp.8-20, 2004.

J. S. Tabb and T. Ueda, Phylogenetic studies on the synaptic vesicle glutamate transport system, J Neurosci, vol.11, issue.6, pp.1822-1828, 1991.

A. Thomas, Depression in older people, Oxford Textbook of old age psychiatry, pp.545-570, 2013.

S. Takamori, M. Holt, K. Stenius, E. A. Lemke, M. Gronborg et al.,

B. Schenck, P. Brugger, S. A. Ringler, B. Muller, F. Rammner et al.,

G. Groot, Y. Mieskes, J. Moriyama, H. Klingauf, J. Grubmuller et al.,

. Jahn, Molecular anatomy of a trafficking organelle, Cell, vol.127, issue.4, pp.831-846, 2006.

S. Takamori, P. Malherbe, C. Broger, and R. Jahn, Molecular cloning and functional characterization of human vesicular glutamate transporter 3, EMBO Rep, vol.3, issue.8, pp.798-803, 2002.

S. Takamori, J. S. Rhee, C. Rosenmund, and R. Jahn, Identification of a vesicular glutamate transporter that defines a glutamatergic phenotype in neurons, Nature, vol.407, issue.6801, pp.189-194, 2000.

S. Takamori, J. S. Rhee, C. Rosenmund, and R. Jahn, Identification of differentiation-associated brain-specific phosphate transporter as a second vesicular glutamate transporter (VGLUT2), J Neurosci, vol.21, issue.22, p.182, 2001.

T. Takeda, Senescence-accelerated mouse (SAM): a biogerontological resource in aging research, Neurobiol Aging, vol.20, issue.2, pp.105-110, 1999.

T. Takeda, Senescence-accelerated mouse (SAM) with special references to neurodegeneration models, SAMP8 and SAMP10 mice, Neurochem Res, vol.34, issue.4, pp.639-659, 2009.

T. Takeda, M. Hosokawa, K. Higuchi, M. Hosono, I. Akiguchi et al., A novel murine model of aging, Senescence-Accelerated Mouse (SAM), Arch Gerontol Geriatr, vol.19, issue.2, pp.185-192, 1994.

T. Takeda, M. Hosokawa, S. Takeshita, M. Irino, K. Higuchi et al.,

K. Yasuhira, H. Hamamoto, K. Shimizu, M. Ishii, and T. Yamamuro, A new murine model of accelerated senescence, Mech Ageing Dev, vol.17, issue.2, pp.183-194, 1981.

N. Takei, I. Nihonmatsu, and H. Kawamura, Age-related decline of acetylcholine release evoked by depolarizing stimulation, Neurosci Lett, vol.101, issue.2, p.182, 1989.

Y. Tamura, K. Ogita, and T. Ueda, A new VGLUT-specific potent inhibitor: pharmacophore of Brilliant Yellow, Neurochem Res, vol.39, issue.1, pp.117-128, 2014.

J. Tanaka, Y. Okuma, K. Tomobe, and Y. Nomura, The age-related degeneration of oligodendrocytes in the hippocampus of the senescence-accelerated mouse (SAM) P8: a quantitative immunohistochemical study, Biol Pharm Bull, vol.28, issue.4, pp.615-618, 2005.

A. Terman and U. T. Brunk, Aging as a catabolic malfunction, Int J Biochem Cell Biol, vol.36, issue.12, pp.2365-2375, 2004.

M. Thoby-brisson, M. Karlen, N. Wu, P. Charnay, J. Champagnat et al., , 2009.

, Genetic identification of an embryonic parafacial oscillator coupling to the preBotzinger complex, Nat Neurosci, vol.12, issue.8, pp.1028-1035

K. Tomobe and Y. Nomura, Neurochemistry, neuropathology, and heredity in SAMP8: a mouse model of senescence, Neurochem Res, vol.34, issue.4, pp.660-669, 2009.

N. Toni, P. A. Buchs, I. Nikonenko, C. R. Bron, and D. Muller, LTP promotes formation of multiple spine synapses between a single axon terminal and a dendrite, Nature, vol.402, issue.6760, pp.421-425, 1999.

R. M. Tordera, S. Totterdell, S. M. Wojcik, N. Brose, N. Elizalde et al., Enhanced anxiety, depressive-like behaviour and impaired recognition memory in mice with reduced expression of the vesicular glutamate transporter 1 (VGLUT1), Eur J Neurosci, vol.25, issue.1, pp.281-290, 2007.

S. Trowbridge, N. Narboux-neme, and P. Gaspar, Genetic models of serotonin (5-HT) depletion: what do they tell us about the developmental role of 5-HT?, Anat Rec (Hoboken), vol.294, issue.10, pp.1615-1623, 2011.

Y. Uchida, S. Sugiura, M. Sone, H. Ueda, and T. Nakashima, Progress and prospects in human genetic research into age-related hearing impairment, Biomed Res Int, p.390601, 2014.

M. Ueno, H. Sakamoto, K. Kanenishi, M. Onodera, I. Akiguchi et al., Ultrastructural and permeability features of microvessels in the hippocampus, cerebellum and pons of senescence-accelerated mice (SAM), Neurobiol Aging, vol.22, issue.3, pp.469-478, 2001.

A. Uezato, J. H. Meador-woodruff, and R. E. Mccullumsmith, Vesicular glutamate transporter mRNA expression in the medial temporal lobe in major depressive disorder, bipolar disorder, and schizophrenia, Bipolar Disord, vol.11, issue.7, p.711, 2009.

A. L. Upton, N. Salichon, C. Lebrand, A. Ravary, R. Blakely et al., Excess of serotonin (5-HT) alters the segregation of ispilateral and contralateral retinal projections in monoamine oxidase A knock-out mice: possible role of 5-HT uptake in retinal ganglion cells during development, J Neurosci, vol.19, issue.16, p.7007, 1999.

J. M. Van-deursen, The role of senescent cells in ageing, Nature, vol.509, 2014.

V. Varga, A. Losonczy, B. V. Zemelman, Z. Borhegyi, G. Nyiri et al., Fast synaptic subcortical control of hippocampal circuits, Science, vol.326, issue.5951, pp.449-453, 2009.

H. Varoqui, M. K. Schafer, H. Zhu, E. Weihe, and J. D. Erickson, Identification of the differentiation-associated Na+/PI transporter as a novel vesicular glutamate transporter expressed in a distinct set of glutamatergic synapses, J Neurosci, vol.22, issue.1, pp.142-155, 2002.

J. C. Viemari, H. Burnet, M. Bevengut, and G. Hilaire, Perinatal maturation of the mouse respiratory rhythm-generator: in vivo and in vitro studies, Eur J Neurosci, vol.17, issue.6, pp.1233-1244, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00285953

E. Vigneault, O. Poirel, M. Riad, J. Prud'homme, S. Dumas et al.,

S. E. Mechawar and . Mestikawy, Distribution of vesicular glutamate transporters in the human brain, Front Neuroanat, vol.9, p.23, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01221681

V. Villar-cervino, C. Rocancourt, A. Menuet, C. D. Silva, P. Wincker et al.,

M. C. Mazan and . Rodicio, A vesicular glutamate transporter in lampreys: cDNA cloning and early expression in the nervous system, J Chem Neuroanat, vol.40, issue.1, pp.71-81, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00486376

G. Vogel, D. Neill, M. Hagler, and D. Kors, A new animal model of endogenous depression: a summary of present findings, Neurosci Biobehav Rev, vol.14, issue.1, pp.85-91, 1990.

A. N. Voisin, O. Mnie-filali, N. Giguere, G. M. Fortin, E. Vigneault et al., Axonal Segregation and Role of the Vesicular Glutamate Transporter VGLUT3 in Serotonin Neurons, Front Neuroanat, vol.10, p.39, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01310490

N. Voituron, Y. Shvarev, C. Menuet, M. Bevengut, C. Fasano et al., Fluoxetine treatment abolishes the in vitro respiratory response to acidosis in neonatal mice, PLoS One, vol.5, issue.10, p.13644, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00625245

N. Volkmann, F. M. Marassi, D. D. Newmeyer, and D. Hanein, The rheostat in the membrane: BCL-2 family proteins and apoptosis, Cell Death Differ, vol.21, issue.2, pp.206-215, 2014.

D. Wallach, T. B. Kang, C. P. Dillon, and D. R. Green, Programmed necrosis in inflammation: Toward identification of the effector molecules, Science, vol.352, 2016.

H. H. Wang, H. L. Li, R. Liu, Y. Zhang, K. Liao et al., , 2010.

, Tau overexpression inhibits cell apoptosis with the mechanisms involving multiple viability-related factors, J Alzheimers Dis, vol.21, issue.1, pp.167-179

J. Z. Wang and Z. H. Wang, Senescence may mediate conversion of tau phosphorylation-induced apoptotic escape to neurodegeneration, Exp Gerontol, vol.68, p.82, 2015.

J. Z. Wang, Z. H. Wang, and Q. Tian, Tau hyperphosphorylation induces apoptotic escape and triggers neurodegeneration in Alzheimer's disease, Neurosci Bull, vol.30, issue.2, pp.359-366, 2014.

L. Wang, Q. Jiang, J. Chu, L. Lin, X. G. Li et al., Expression of Tau40 induces activation of cultured rat microglial cells, PLoS One, vol.8, issue.10, p.76057, 2013.

W. Wang, J. K. Tiwari, S. R. Bradley, R. V. Zaykin, and G. B. Richerson, , 2001.

, Acidosis-stimulated neurons of the medullary raphe are serotonergic, J Neurophysiol, vol.85, issue.5, pp.2224-2235

Y. Wei, S. Sinha, and B. Levine, Dual role of JNK1-mediated phosphorylation of Bcl-2 in autophagy and apoptosis regulation, Autophagy, vol.4, issue.7, pp.949-951, 2008.

M. F. Weiner and A. M. Lipton, Clinical Manual of Alzheimer disease and others dementias, American Psychiatric Association, 2012.

M. J. West, P. D. Coleman, D. G. Flood, and J. C. Troncoso, Differences in the pattern of hippocampal neuronal loss in normal ageing and Alzheimer's disease, Lancet, vol.344, issue.8925, pp.769-772, 1994.

D. Westphal, G. Dewson, P. E. Czabotar, and R. M. Kluck, Molecular biology of Bax and Bak activation and action, Biochim Biophys Acta, vol.1813, issue.4, pp.521-531, 2011.

N. R. Wilson, J. Kang, E. V. Hueske, T. Leung, H. Varoqui et al., Presynaptic regulation of quantal size by the vesicular glutamate transporter VGLUT1, J Neurosci, vol.25, issue.26, pp.6221-6234, 2005.

E. Wirawan, T. Vanden-berghe, S. Lippens, P. Agostinis, and P. Vandenabeele, , 2012.

, Autophagy: for better or for worse, Cell Res, vol.22, issue.1, pp.43-61

H. M. Wisniewski and J. Wegiel, Spatial relationships between astrocytes and classical plaque components, Neurobiol Aging, vol.12, issue.5, pp.593-600, 1991.

S. Wittmeier, G. Song, J. Duffin, and C. S. Poon, Pacemakers handshake synchronization mechanism of mammalian respiratory rhythmogenesis, Proc Natl Acad Sci U S A, vol.105, issue.46, pp.18000-18005, 2008.

S. M. Wojcik, J. S. Rhee, E. Herzog, A. Sigler, R. Jahn et al.,

. Rosenmund, An essential role for vesicular glutamate transporter 1 (VGLUT1) in postnatal development and control of quantal size, Proc Natl Acad Sci U S A, vol.101, issue.18, 2004.

C. S. Woolley, N. G. Weiland, B. S. Mcewen, and P. A. Schwartzkroin, , 1997.

, Estradiol increases the sensitivity of hippocampal CA1 pyramidal cells to NMDA receptor-mediated synaptic input: correlation with dendritic spine density, J Neurosci, vol.17, issue.5, pp.1848-1859

H. Wootz, A. Enjin, A. Wallen-mackenzie, D. Lindholm, and K. Kullander, , 2010.

, Reduced VGLUT2 expression increases motor neuron viability in Sod1(G93A) mice, Neurobiol Dis, vol.37, issue.1, pp.58-66

Z. Xie and D. J. Klionsky, Autophagosome formation: core machinery and adaptations, Nat Cell Biol, vol.9, issue.10, pp.1102-1109, 2007.

H. Yagi, A. Akiguchi, N. Ohta, M. Yagi, T. Hosokawa et al., , 1998.

H. Yagi, M. Irino, T. Matsushita, S. Katoh, M. Umezawa et al., Spontaneous spongy degeneration of the brain stem in SAM-P/8 mice, a newly developed memory-deficient strain, J Neuropathol Exp Neurol, vol.48, issue.5, pp.577-590, 1989.

S. Yamada, S. Uchida, T. Ohkura, R. Kimura, M. Yamaguchi et al.,

. Yamamoto, Alterations in calcium antagonist receptors and calcium content in senescent brain and attenuation by nimodipine and nicardipine, J Pharmacol Exp Ther, vol.277, issue.2, pp.721-727, 1996.

T. Yamaguchi, W. Sheen, and M. Morales, Glutamatergic neurons are present in the rat ventral tegmental area, Eur J Neurosci, vol.25, pp.106-118, 2007.

S. Yang, H. Qiao, L. Wen, W. Zhou, and Y. Zhang, D-serine enhances impaired long-term potentiation in CA1 subfield of hippocampal slices from aged senescenceaccelerated mouse prone/8, Neurosci Lett, vol.379, issue.1, pp.7-12, 2005.

Z. Yang and D. J. Klionsky, Mammalian autophagy: core molecular machinery and signaling regulation, Curr Opin Cell Biol, vol.22, issue.2, pp.124-131, 2010.

M. M. Young, Y. Takahashi, O. Khan, S. Park, T. Hori et al.,

D. Hu, J. Zhang, M. Kester, and H. G. Wang, Autophagosomal membrane serves as platform for intracellular death-inducing signaling complex (iDISC)-mediated caspase-8 activation and apoptosis, J Biol Chem, vol.287, issue.15, pp.12455-12468, 2012.

S. Yousefi, R. Perozzo, I. Schmid, A. Ziemiecki, T. Schaffner et al., Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis, Nat Cell Biol, vol.8, issue.10, pp.1124-1132, 2006.

L. Yu, A. Alva, H. Su, P. Dutt, E. Freundt et al., Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8, Science, vol.304, issue.5676, pp.1500-1502, 2004.

S. W. Yu, S. H. Baek, R. T. Brennan, C. J. Bradley, S. K. Park et al.,

E. K. Lookingland, H. Kim, J. L. Lee, S. W. Goudreau, and . Kim, Autophagic death of adult hippocampal neural stem cells following insulin withdrawal, Stem Cells, vol.26, issue.10, 2008.

X. Yu and S. He, The interplay between human herpes simplex virus infection and the apoptosis and necroptosis cell death pathways, Virol J, vol.13, p.77, 2016.

Z. Y. Yuan, P. Gu, L. Liu, Y. Y. Wang, J. Liu et al.,

M. W. Ma and . Wang, Neuroprotective effects of enriched environment in MPTPtreated SAMP8 mice, Neurosci Lett, vol.454, issue.1, pp.6-10, 2009.

E. Zalckvar, H. Berissi, M. Eisenstein, and A. Kimchi, Phosphorylation of, 2009.

, Beclin 1 by DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL, Autophagy, vol.5, issue.5, pp.720-722

E. Zalckvar, H. Berissi, L. Mizrachy, Y. Idelchuk, I. Koren et al., DAP-kinase-mediated phosphorylation on the BH3 domain of beclin 1 promotes dissociation of beclin 1 from Bcl-XL and induction of autophagy, EMBO Rep, vol.10, issue.3, pp.285-292, 2009.

J. Zhang and P. A. Ney, Role of BNIP3 and NIX in cell death, autophagy, and mitophagy, Cell Death Differ, vol.16, issue.7, pp.939-946, 2009.

Q. Zhang, H. Ding, W. Li, Z. Fan, A. Sun et al., Senescence accelerated mouse strain is sensitive to neurodegeneration induced by mild impairment of oxidative metabolism, Brain Res, vol.1264, pp.111-118, 2009.

W. Zhang, A. Barnbrock, S. Gajic, A. Pfeiffer, and B. Ritter, Differential ontogeny of GABA(B)-receptor-mediated pre-and postsynaptic modulation of GABA and glycine transmission in respiratory rhythm-generating network in mouse, J Physiol, vol.540, pp.435-446, 2002.

X. H. Zhao, Y. Kitamura, and Y. Nomura, Age-related changes in NMDAinduced [3H]acetylcholine release from brain slices of senescence-accelerated mouse, 1992.

, Int J Dev Neurosci, vol.10, issue.2, pp.121-129

W. Zhou and J. Yuan, Necroptosis in health and diseases, Semin Cell Dev Biol, vol.35, pp.14-23, 2014.

Y. Zhu, L. Zhao, L. Liu, P. Gao, W. Tian et al., , 2010.

, Beclin 1 cleavage by caspase-3 inactivates autophagy and promotes apoptosis, Protein Cell, vol.1, issue.5, pp.468-477

J. Zimmermann, M. A. Herman, and C. Rosenmund, Co-release of glutamate and GABA from single vesicles in GABAergic neurons exogenously expressing VGLUT3, Front Synaptic Neurosci, vol.7, p.16, 2015.

M. Zink, B. Vollmayr, P. J. Gebicke-haerter, and F. A. Henn, Reduced expression of glutamate transporters vGluT1, EAAT2 and EAAT4 in learned helpless rats, an animal model of depression, Neuropharmacology, vol.58, issue.2, pp.465-473, 2010.