D. Bouhassira, M. Lanteri-minet, N. Attal, B. Laurent, and C. Touboul, Prevalence of chronic pain with neuropathic characteristics in the general population, Pain, vol.136, pp.380-387, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00326893

M. Ospina and C. Harstall, Prevalence of Chronic Pain: an Overview, Health Technology Assessment, 2002.

P. F. Verhaak, J. J. Kerssens, J. Dekker, M. J. Sorbi, and J. M. Bensing, Prevalence of chronic benign pain disorder among adults: a review of the literature, Pain, vol.77, pp.231-239, 1998.

H. Breivik, B. Collett, V. Ventafridda, R. Cohen, and D. Gallacher, Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment, Eur J Pain, vol.10, pp.287-333, 2006.

N. B. Finnerup, S. H. Sindrup, and T. S. Jensen, The evidence for pharmacological treatment of neuropathic pain, Pain, vol.150, pp.573-581, 2010.

M. S. Mercier, D. Lodge, G. Fang, C. S. Nicolas, V. J. Collett et al., Characterisation of an mGlu8 receptor-selective agonist and antagonist in the lateral and medial perforant path inputs to the dentate gyrus, Neuropharmacology, vol.67, pp.294-303, 2013.

H. Y. Zhou, H. M. Zhang, S. R. Chen, and H. L. Pan, Increased nociceptive input rapidly modulates spinal GABAergic transmission through endogenously released glutamate, J Neurophysiol, vol.97, pp.871-882, 2007.

A. E. Kingston, P. L. Ornstein, R. A. Wright, B. G. Johnson, N. G. Mayne et al., LY341495 is a nanomolar potent and selective antagonist of group II metabotropic glutamate receptors, Neuropharmacology, vol.37, pp.1-12, 1998.

Y. C. Liang, C. C. Huang, and K. S. Hsu, Characterization of long-term potentiation of primary afferent transmission at trigeminal synapses of juvenile rats: essential role of subtype 5 metabotropic glutamate receptors, Pain, vol.114, pp.417-428, 2005.

M. E. Fundytus, J. Ritchie, and T. J. Coderre, Attenuation of morphine withdrawal symptoms by subtype-selective metabotropic glutamate receptor antagonists, Br J Pharmacol, vol.120, pp.1015-1020, 1997.

E. Palazzo, I. Marabese, V. De-novellis, P. Oliva, F. Rossi et al., Metabotropic and NMDA glutamate receptors participate in the cannabinoid-induced antinociception, Neuropharmacology, vol.40, pp.319-326, 2001.

E. Palazzo, V. De-novellis, I. Marabese, D. Cuomo, F. Rossi et al., Interaction between vanilloid and glutamate receptors in the central modulation of nociception, European journal of pharmacology, vol.439, pp.69-75, 2002.

Y. H. Jin, M. Takemura, A. Furuyama, and N. Yonehara, Peripheral glutamate receptors are required for hyperalgesia induced by capsaicin, Pain Res Treat, p.915706, 2012.

S. Dolan, M. D. Gunn, L. Biddlestone, and A. M. Nolan, The selective metabotropic glutamate receptor 7 allosteric agonist AMN082 inhibits inflammatory pain-induced and incisioninduced hypersensitivity in rat, Behav Pharmacol, vol.20, pp.596-604, 2009.

M. Maj, V. Bruno, Z. Dragic, R. Yamamoto, G. Battaglia et al., PHCCC, a positive allosteric modulator of mGluR4: characterization, mechanism of action, and neuroprotection, Neuropharmacology, vol.45, pp.895-906, 2003.

M. J. Marino, D. L. Williams, J. O'brien, J. A. Valenti, O. Mcdonald et al., Allosteric modulation of group III metabotropic glutamate receptor 4: a potential approach to Parkinson's disease treatment, Proc Natl Acad Sci U S A, vol.100, pp.13668-13673, 2003.

C. M. Niswender, K. A. Johnson, C. D. Weaver, C. K. Jones, Z. Xiang et al., Discovery, characterization, and antiparkinsonian effect of novel positive allosteric modulators of metabotropic glutamate receptor 4, Mol pharmacol, vol.74, pp.1345-1358, 2008.

S. Celanire and B. Campo, Recent advances in the drug discovery of metabotropic glutamate receptor 4 (mGluR4) activators for the treatment of CNS and non-CNS disorders, Expert Opin Drug Discov, vol.7, pp.261-280, 2012.

R. M. Duvoisin, T. Pfankuch, J. M. Wilson, J. Grabell, V. Chhajlani et al., Acute pharmacological modulation of mGluR8 reduces measures of anxiety, Behavioural brain research, vol.212, pp.168-173, 2010.

R. M. Duvoisin, L. Villasana, M. J. Davis, D. G. Winder, and J. Raber, Opposing roles of mGluR8 in measures of anxiety involving non-social and social challenges, Behav Brain Res, vol.221, pp.50-54, 2011.

K. Mitsukawa, R. Yamamoto, S. Ofner, J. Nozulak, O. Pescott et al., A selective metabotropic glutamate receptor 7 agonist: activation of receptor signaling via an allosteric site modulates stress parameters in vivo, Proc Natl Acad Sci, vol.102, pp.18712-18717, 2005.

M. Osikowicz, J. Mika, W. Makuch, and B. Przewlocka, Glutamate receptor ligands attenuate allodynia and hyperalgesia and potentiate morphine effects in a mouse model of neuropathic pain, Pain, vol.139, pp.117-126, 2008.

H. Wang, W. Jiang, R. Yang, and Y. Li, Spinal metabotropic glutamate receptor 4 is involved in neuropathic pain, Neuroreport, vol.22, issue.5, pp.244-248, 2011.

S. Rizzo, S. J. Leonard, S. K. Gilbert, A. Dollings, P. Smith et al., The metabotropic glutamate receptor 7 allosteric modulator AMN082: a monoaminergic agent in disguise?, J Pharmacol Exp Ther, vol.338, issue.1, pp.345-352, 2011.

, *The orally active and brain penetrant mGlu7 PAM AMN082 is a very useful ligand to investigate the function receptor. However, this study clearly shows that this molecule is rapidly metabolized in vivo (t1/2 < 1min) and that its major metabolite is a potent inhibitor of monoamine transporters, This suggests that in vivo activity of AMN082 may involve other targets than mGlu7

G. Suzuki, N. Tsukamoto, H. Fushiki, A. Kawagishi, M. Nakamura et al., In vitro pharmacological characterization of novel isoxazolopyridone derivatives as allosteric metabotropic glutamate receptor 7 antagonists, J Pharmacol Exp Ther, vol.323, pp.147-156, 2007.

E. Palazzo, I. Marabese, L. Luongo, S. Boccella, G. Bellini et al., Effects of a metabotropic glutamate receptor subtype 7 negative allosteric modulator in the periaqueductal grey on pain responses and rostral ventromedial medulla cell activity in rat, Molecular pain, vol.9, p.44, 2013.

C. M. Niswender, K. A. Johnson, N. R. Miller, J. E. Ayala, Q. Luo et al., Context-dependent pharmacology exhibited by negative allosteric modulators of metabotropic glutamate receptor 7, Mol Pharmacol, vol.77, pp.459-468, 2010.

M. Kalinichev, M. Rouillier, F. Girard, I. Royer-urios, B. Bournique et al., ADX71743, a potent and selective negative allosteric modulator of metabotropic glutamate receptor 7 (mGlu7): in vitro and in vivo characterization, J Pharmacol Exp Ther, vol.344, pp.624-636, 2013.

, *In this study, the authors describe a very complete characterization of a potent, brain penetrant and selective mGlu7 NAM ADX71743 both in vitro and in vivo in many behavioural models

C. E. Gee, D. Peterlik, C. Neuhauser, R. Bouhelal, K. Kaupmann et al., Blocking metabotropic glutamate receptor subtype 7 (mGlu7) via the Venus flytrap domain (VFTD) inhibits amygdala plasticity, stress, and anxiety-related behavior, J Biol Chem, vol.289, pp.10975-10987, 2014.

, This study describes XAP044, a selective and brain penetrant mGlu7 NAM with anti-stress, antidepressant-and anxiolytic-like efficacy in vivo, able to modulate synaptic plasticity within amygdala and which likely targets an allosteric pocket located within the extracellular domain of the receptor contrary to other mGluRs NAMs described so far

R. Pekhletski, R. Gerlai, L. S. Overstreet, X. P. Huang, N. Agopyan et al., Impaired cerebellar synaptic plasticity and motor performance in mice lacking the mGluR4 subtype of metabotropic glutamate receptor, J Neurosci, vol.16, pp.6364-6373, 1996.

R. Gerlai, J. C. Roder, and D. R. Hampson, Altered spatial learning and memory in mice lacking the mGluR4 subtype of metabotropic glutamate receptor, Behav Neurosci, vol.112, pp.525-532, 1998.

G. Sansig, T. J. Bushell, V. R. Clarke, A. Rozov, N. Burnashev et al., Increased seizure susceptibility in mice lacking metabotropic glutamate receptor 7, J Neurosci, vol.21, pp.8734-8745, 2001.

J. F. Cryan, P. H. Kelly, H. C. Neijt, G. Sansig, P. J. Flor et al., Antidepressant and anxiolytic-like effects in mice lacking the group III metabotropic glutamate receptor mGluR7, Eur J Neurosci, vol.17, pp.2409-2417, 2003.

A. M. Linden, B. G. Johnson, S. C. Peters, H. E. Shannon, M. Tian et al., Increased anxiety-related behavior in mice deficient for metabotropic glutamate 8 (mGlu8) receptor, Neuropharmacology, vol.43, pp.251-259, 2002.

R. M. Duvoisin, C. Zhang, T. F. Pfankuch, H. O&apos;connor, J. Gayet-primo et al., Increased measures of anxiety and weight gain in mice lacking the group III metabotropic glutamate receptor mGluR8, Eur J Neurosci, vol.22, pp.425-436, 2005.

M. Fendt, S. Imobersteg, D. Peterlik, F. Chaperon, C. Mattes et al., amygdala-dependent behavior and physiology, vol.72, pp.215-223, 2013.

, * Combining pharmacological and electrophysiological approaches with mGlu7 and mGlu8 KO mice, the authors investigated the roles of mGlu7 and mGlu8 in amygdala-dependent behavior and synaptic plasticity. It revealed the different roles for mGlu7 and mGlu8 in amygdala synaptic transmission

R. M. Govea, S. Zhou, and S. M. Carlton, Group III metabotropic glutamate receptors and transient receptor potential vanilloid 1 co-localize and interact on nociceptors, Neuroscience, vol.217, pp.130-139, 2012.

A. J. Todd, Neuronal circuitry for pain processing in the dorsal horn, Nature reviews Neuroscience, vol.11, pp.823-836, 2010.

J. J. Azkue, M. Murga, O. Fernandez-capetillo, J. M. Mateos, I. Elezgarai et al., Immunoreactivity for the group III metabotropic glutamate receptor subtype mGluR4a in the superficial laminae of the rat spinal dorsal horn, J Comp Neurol, vol.430, pp.448-457, 2001.

H. Ohishi, C. Akazawa, R. Shigemoto, S. Nakanishi, and N. Mizuno, Distributions of the mRNAs for L-2-amino-4-phosphonobutyrate-sensitive metabotropic glutamate receptors, mGluR4 and mGluR7, in the rat brain, J Comp Neurol, vol.360, pp.555-570, 1995.

H. Ohishi, S. Nomura, Y. Q. Ding, R. Shigemoto, E. Wada et al., Presynaptic localization of a metabotropic glutamate receptor, mGluR7, in the primary afferent neurons: an immunohistochemical study in the rat, Neurosci Lett, vol.202, pp.85-88, 1995.

S. M. Carlton and G. L. Hargett, Colocalization of metabotropic glutamate receptors in rat dorsal root ganglion cells, J Comp Neurol, vol.501, pp.780-789, 2007.

A. Valerio, M. Paterlini, M. Boifava, M. Memo, and P. Spano, Metabotropic glutamate receptor mRNA expression in rat spinal cord, Neuroreport, vol.8, pp.2695-2699, 1997.

R. M. Duvoisin, C. Zhang, and K. Ramonell, A novel metabotropic glutamate receptor expressed in the retina and olfactory bulb, J Neurosci, vol.15, pp.3075-3083, 1995.

C. D. Mills, K. M. Johnson, and C. E. Hulsebosch, Role of group II and group III metabotropic glutamate receptors in spinal cord injury, Exp Neurol, vol.173, pp.153-167, 2002.

L. Neto, F. Schadrack, J. Berthele, A. Zieglgansberger, W. Tolle et al., Differential distribution of metabotropic glutamate receptor subtype mRNAs in the thalamus of the rat, Brain research, vol.854, pp.93-105, 2000.

V. Neugebauer, Chapter 11 Subcortical processing of nociceptive information: basal ganglia and amygdala, Handb Clin Neurol, vol.81, pp.141-158, 2006.

J. S. Han, G. C. Bird, and V. Neugebauer, Enhanced group III mGluR-mediated inhibition of painrelated synaptic plasticity in the amygdala, Neuropharmacology, vol.46, pp.918-926, 2004.

M. J. Millan, Descending control of pain, Prog Neurobiol, vol.66, pp.355-474, 2002.

S. Maione, E. Palazzo, V. De-novellis, L. Stella, J. Leyva et al., Metabotropic glutamate receptors modulate serotonin release in the rat periaqueductal gray matter, Naunyn Schmiedebergs Arch Pharmacol, vol.358, pp.411-417, 1998.

E. Palazzo, V. De-novellis, F. Rossi, and S. Maione, Supraspinal metabotropic glutamate receptor subtype 8: a switch to turn off pain, Amino Acids, vol.46, pp.1441-1448, 2014.

E. Palazzo, Y. Fu, J. G. Maione, S. Neugebauer, and V. , Group III mGluR7 and mGluR8 in the amygdala differentially modulate nocifensive and affective pain behaviors, vol.55, pp.537-545, 2008.

E. Palazzo, I. Marabese, M. Soukupova, L. Luongo, S. Boccella et al., Metabotropic glutamate receptor subtype 8 in the amygdala modulates thermal threshold, neurotransmitter release, and rostral ventromedial medulla cell activity in inflammatory pain, J Neurosci, vol.31, pp.4687-4697, 2011.

A. Dobi, S. B. Sartori, D. Busti, H. Van-der-putten, N. Singewald et al., Neural substrates for the distinct effects of presynaptic group III metabotropic glutamate receptors on extinction of contextual fear conditioning in mice, Neuropharmacology, vol.66, pp.274-289, 2013.

J. P. Pin, C. De-colle, A. S. Bessis, and F. Acher, New perspectives for the development of selective metabotropic glutamate receptor ligands, Eur J Pharmacol, vol.375, pp.277-294, 1999.

D. D. Schoepp, D. E. Jane, and J. A. Monn, Pharmacological agents acting at subtypes of metabotropic glutamate receptors, Neuropharmacology, vol.38, pp.1431-1476, 1999.