H. A. Quigley and A. T. Broman, The number of people with glaucoma worldwide in 2010 and 2020, Br. J. Ophthalmol, vol.90, pp.262-267, 2006.

C. Cedrone, R. Mancino, A. Cerulli, M. Cesareo, and C. Nucci, Epidemiology of primary glaucoma: prevalence, incidence, and blinding effects, Prog. Brain. Res, vol.173, pp.3-14, 2008.

A. Sommer, Intraocular pressure and glaucoma, Am. J. Ophthalmol, vol.107, pp.186-188, 1989.

M. A. Kass, D. K. Heuer, E. J. Higginbotham, C. A. Johnson, and J. L. Keltner, The Ocular Hypertension Treatment Study: a randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma, Arch. Ophthalmol, vol.120, pp.701-713, 2002.

O. Y. Tektas and E. Lütjen-drecoll, Structural changes of the trabecular meshwork in different kinds of glaucoma, Exp. Eye Res, vol.88, pp.769-775, 2009.

J. Alvarado, C. Murphy, and R. Juster, Trabecular meshwork cellularity in primary open-angle glaucoma and non-glaucomatous normals, Ophthalmology, vol.91, pp.564-579, 1984.

J. W. Rohen, E. Lutjen-drecoll, C. Flugel, M. Meyer, and I. Grierson, Ultrastructure of the trabecular meshwork in untreated cases of primary openangle glaucoma, Exp. Eye Res, vol.56, pp.683-692, 1993.

I. Grierson and R. C. Howes, Age-related depletion of the cell population in the human trabecular meshwork, Eye, vol.1, pp.204-210, 1987.

P. Hamard, F. Valtot, P. Sourdille, F. Bourles-dagonet, and C. Baudouin, Confocal microscopic examination of trabecular meshwork removed during ab externo trabeculectomy, Br. J. Ophthalmol, vol.86, pp.1046-1052, 2002.

J. Baleriola, J. Garcia-feijoo, J. M. Martinez-de-la-casa, A. Fernandez-cruz, and E. J. De-la-rosa, Apoptosis in the trabecular meshwork of glaucomatous patients, Mol. Vis, vol.14, pp.1513-1516, 2008.

K. Balabanian, B. Lagane, S. Infantino, K. Y. Chow, and J. Harriague, The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes, J. Biol. Chem, vol.280, pp.35760-35766, 2005.

J. M. Burns, B. C. Summers, Y. Wang, A. Melikian, and R. Berahovich, A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development, J. Exp. Med, vol.203, pp.2201-2213, 2006.

M. Thelen and S. Thelen, CXCR7, CXCR4 and CXCL12: an eccentric trio?, J. Neuroimmunol, vol.198, pp.9-13, 2008.

M. Li and R. M. Ransohoff, Multiple roles of chemokine CXCL12 in the central nervous system: a migration from immunology to neurobiology, Prog. Neurobiol, vol.84, pp.116-131, 2008.

W. Rostene, P. Kitabgi, and S. M. Parsadaniantz, Chemokines: a new class of neuromodulator?, Nat. Rev. Neurosci, vol.8, pp.895-903, 2007.

A. Muller, B. Homey, H. Soto, N. Ge, and D. Catron, Involvement of chemokine receptors in breast cancer metastasis, Nature, vol.410, pp.50-56, 2001.

T. Lapidot, A. Dar, and O. Kollet, How do stem cells find their way home?, Blood, vol.106, pp.1901-1910, 2005.

B. R. Son, L. A. Marquez-curtis, M. Kucia, M. Wysoczynski, and A. R. Turner, Migration of bone marrow and cord blood mesenchymal stem cells in vitro is regulated by stromal-derived factor-1-CXCR4 and hepatocyte growth factor-c-met axes and involves matrix metalloproteinases, Stem Cells, vol.24, pp.1254-1264, 2006.

I. J. Crane, C. A. Wallace, S. Mckillop-smith, and J. V. Forrester, CXCR4 receptor expression on human retinal pigment epithelial cells from the blood-retina barrier leads to chemokine secretion and migration in response to stromal cellderived factor 1 alpha, J. Immunol, vol.165, pp.4372-4378, 2000.

I. A. Bhutto, D. S. Mcleod, C. Merges, T. Hasegawa, and G. A. Lutty, Localisation of SDF-1 and its receptor CXCR4 in retina and choroid of aged human eyes and in eyes with age related macular degeneration, Br. J. Ophthalmol, vol.90, pp.906-910, 2006.

T. Bourcier, T. Berbar, S. Paquet, N. Rondeau, and F. Thomas, Characterization and functionality of CXCR4 chemokine receptor and SDF-1 in human corneal fibroblasts, Mol. Vis, vol.9, pp.96-102, 2003.

S. J. Curnow, K. Wloka, J. M. Faint, N. Amft, and C. M. Cheung, Topical glucocorticoid therapy directly induces up-regulation of functional CXCR4 on primed T lymphocytes in the aqueous humor of patients with uveitis, J. Immunol, vol.172, pp.7154-7161, 2004.

G. A. Mcquibban, J. H. Gong, E. M. Tam, C. A. Mcculloh, and C. , Inflammation dampened by gelatinase A cleavage of monocyte chemoattractant protein-3, Science, vol.289, pp.1202-1206, 2000.

H. Zhang, A. Trivedi, J. U. Lee, M. Lohela, and S. M. Lee, Matrix metalloproteinase-9 and stromal cell-derived factor-1 act synergistically to support migration of blood-borne monocytes into the injured spinal cord, J. Neurosci, vol.9, pp.143-149, 2011.

G. A. Mcquibban, G. S. Butler, J. H. Gong, L. Bendall, and C. Power, Matrix metalloproteinase activity inactivates the CXC chemokine stromal cell-derived factor-1, J. Biol. Chem, vol.276, pp.43503-43508, 2001.

A. Valenzuela-fernandez, T. Planchenault, F. Baleux, I. Staropoli, and K. Le-barillec, Leukocyte elastase negatively regulates Stromal cell-derived factor-1 (SDF-1)/CXCR4 binding and functions by amino-terminal processing of SDF-1 and CXCR4, J. Biol. Chem, vol.277, pp.15677-15689, 2002.
URL : https://hal.archives-ouvertes.fr/pasteur-00166872

K. Zhang, G. A. Mcquibban, C. Silva, G. S. Butler, and J. B. Johnston, HIVinduced metalloproteinase processing of the chemokine stromal cell derived factor-1 causes neurodegeneration, Nat. Neurosci, vol.6, pp.1064-1071, 2003.

D. Vergote, G. S. Butler, M. Ooms, J. H. Cox, and C. Silva, Proteolytic processing of SDF-1alpha reveals a change in receptor specificity mediating HIV-associated neurodegeneration, Proc. Natl. Acad. Sci. U.S.A, vol.103, pp.19182-19187, 2006.

J. Medina, Discovery and Development of a CXCR3 antagonist T487 as therapy for Th1-mediated immune disorders, 29th National Medicinal Chemistry Symposium, 2004.

C. E. Heise, A. Pahuja, S. C. Hudson, M. S. Mistry, and A. L. Putnam, Pharmacological characterization of CXC chemokine receptor 3 ligands and a small molecule antagonist, J. Pharmacol. Exp. Ther, vol.313, pp.1263-1271, 2005.

G. A. Donzella, D. Schols, S. W. Lin, J. A. Esté, and K. A. Nagashima, AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 coreceptor, Nat. Med, vol.4, pp.72-77, 1988.

I. H. Pang, D. L. Shade, A. F. Clark, H. T. Steely, and L. Desantis, Preliminary characterization of a transformed cell strain derived from human trabecular meshwork, Curr. Eye. Res, vol.13, pp.51-63, 1994.

E. Garcia-valenzuela, S. Shareef, J. Walsh, and S. C. Sharma, Programmed cell death of retinal ganglion cells during experimental glaucoma, Exp. Eye. Res, vol.61, pp.33-44, 1995.

P. Hamard, C. Blondin, C. Debbash, J. M. Warnet, and C. Baudouin, In vitro effects of preserved and unpreserved antiglaucoma drugs on apoptotic marker expression by human trabecular cells, Graefes Arch. Clin. Exp. Ophthalmol, vol.241, pp.1037-1043, 2003.

E. Borenfreund and J. A. Puerner, Toxicity determined in vitro by morphological alterations and neutral red absorption, Toxicol. Lett, vol.24, pp.119-124, 1985.

C. Debbasch, F. Brignole, P. J. Pisella, J. M. Warnet, and P. Rat, Quaternary ammoniums and other preservatives' contribution in oxidative stress and apoptosis on Chang conjunctival cells, Invest. Ophthalmol. Vis. Sci, vol.42, pp.642-652, 2001.

R. F. Jones and D. M. Maurice, New methods of measuring the rate of aqueous flow in man with fluorescein, Exp. Eye. Res, vol.5, pp.208-220, 1966.

M. E. Yablonski, T. J. Zimmerman, S. R. Waltman, and B. Becker, A fluorophotometric study of the effect of topical timolol on aqueous humor dynamics, Exp. Eye. Res, vol.25, pp.135-142, 1978.

Z. Lu, D. R. Overby, P. A. Scott, T. F. Freddo, and H. Gong, The mechanism of increasing outflow facility by rho-kinase inhibition with Y-27632 in bovine eyes, Exp. Eye. Res, vol.86, pp.271-281, 2008.

Y. Zhang, C. B. Toris, Y. Liu, W. Ye, and H. Gong, Morphological and hydrodynamic correlates in monkey eyes with laser induced glaucoma, Exp. Eye. Res, vol.89, pp.748-756, 2009.
DOI : 10.1016/j.exer.2009.06.015

B. B. Thomas, M. J. Seiler, S. R. Sadda, P. J. Coffey, and R. B. Aramant, Optokinetic test to evaluate visual acuity of each eye independently, J. Neurosci. Methods, vol.138, pp.7-13, 2004.

W. Rostène, M. M. Dansereau, D. Godefroy, J. Van-steenwinckel, R. Goazio et al., Neurochemokines: a ménage a trois providing new insights on the functions of chemokines in the central nervous system, J. Neurochem, vol.118, pp.680-694, 2011.

X. Zhao, K. E. Ramsey, D. A. Stephan, and P. Russell, Gene and protein expression changes in human trabecular meshwork cells treated with transforming growth factor-beta, Invest. Ophthalmol. Vis. Sci, vol.45, pp.4023-4034, 2004.
DOI : 10.1167/iovs.04-0535

URL : https://iovs.arvojournals.org/data/journals/iovs/932923/z7g01104004023.pdf

M. E. Sherwood, T. M. Richardson, and D. L. Epstein, Phagocytosis by trabecular meshwork cells: Sequence of events in cats and monkeys, Exp. Eye Res, vol.46, pp.881-895, 1988.

M. R. Hernandez, B. I. Weinstein, J. Schwartz, R. R. Gordon, and G. G. , Human trabecular meshwork cells in culture: Morphology and extracellular matrix components, Invest. Ophthalmol. Vis. Sci, vol.28, pp.1655-1660, 1987.

T. S. Acott and M. J. Kelley, Extracellular matrix in the trabecular meshwork, Exp. Eye Res, vol.86, pp.543-561, 2008.
DOI : 10.1016/j.exer.2008.01.013

URL : http://europepmc.org/articles/pmc2376254?pdf=render

R. C. Tripathi, J. Li, W. F. Chan, and B. J. Tripathi, Aqueous humor in glaucomatous eyes contains an increased level of TGF-beta 2, Exp. Eye. Res, vol.59, pp.723-727, 1994.
DOI : 10.1006/exer.1994.1158

H. Sawada, T. Fukuchi, T. Tanaka, and H. Abe, Tumor necrosis factor-alpha concentrations in the aqueous humor of patients with glaucoma, Invest. Ophthalmol. Vis. Sci, vol.51, pp.903-906, 2010.

F. Pujol, P. Kitabgi, and H. Boudin, The chemokine SDF-1 differentially regulates axonal elongation and branching in hippocampal neurons, J. Cell. Sci, vol.118, pp.1071-1080, 2005.
DOI : 10.1242/jcs.01694

URL : http://jcs.biologists.org/content/118/5/1071.full.pdf

F. Barbieri, A. Bajetto, R. Stumm, A. Pattarozzi, and C. Porcile, CXC receptor and chemokine expression in human meningioma: SDF1/CXCR4 signaling activates ERK1/2 and stimulates meningioma cell proliferation, Ann. N.Y. Acad. Sci, vol.1090, pp.332-343, 2006.

M. Z. Khan, R. Brandimarti, S. Shimizu, J. Nicolai, and E. Crowe, The chemokine CXCL12 promotes survival of postmitotic neurons by regulating Rb protein, Cell Death Differ, vol.15, pp.1663-1672, 2008.
DOI : 10.1038/cdd.2008.95

URL : https://www.nature.com/articles/cdd200895.pdf

L. Lasagni, M. Francalanci, F. Annunzuato, E. Lazzeri, and S. Giannini, An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, Mig, and I-TAC, and acts as functional receptor for platelet factor 4, J. Exp. Med, vol.197, pp.1537-1549, 2003.

Y. Zhu, D. Vergote, C. Pardo, F. Noorbakhsh, and J. C. Mcarthur, CXCR3 activation by lentivirus infection suppresses neuronal autophagy: neuroprotective effects of antiretroviral therapy, FASEB J, vol.23, pp.2928-2941, 2009.
DOI : 10.1096/fj.08-128819

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2796902/pdf

L. A. Jopling, G. F. Watt, S. Fisher, H. Birch, and S. Coggon, Analysis of the pharmacokinetic/pharmacodynamic relationship of a small molecule CXCR3 antagonist, NBI-74330, using a murine CXCR3 internalization assay, Br. J. Pharmacol, vol.152, pp.1260-1271, 2007.

D. Verzijl, S. Storelli, D. J. Scholten, L. Bosch, and T. A. Reinhart, Noncompetitive antagonism and inverse agonism as mechanism of action of nonpeptidergic antagonists at primate and rodent CXCR3 chemokine receptors, J. Pharmacol. Exp. Ther, vol.325, pp.544-555, 2008.
DOI : 10.1124/jpet.107.134783

URL : http://jpet.aspetjournals.org/content/325/2/544.full.pdf

E. J. Van-wanrooij, S. C. Jager, T. Van-es, P. De-vos, and H. L. Birch, CXCR3 antagonist NBI-74330 attenuates atherosclerotic plaque formation in LDL receptor-deficient mice, Arterioscler. Thromb. Vasc. Biol, vol.28, pp.251-257, 2008.

C. Liu, D. Luo, B. A. Reynolds, G. Meher, and A. R. Katritzky, Chemokine receptor CXCR3 promotes growth of glioma, Carcinogenesis, vol.32, pp.129-137, 2011.
DOI : 10.1093/carcin/bgq224

URL : http://europepmc.org/articles/pmc3026840?pdf=render

J. Danias, F. Shen, M. Kavalarakis, B. Chen, and D. Goldblum, Characterization of retinal damage in the episcleral vein cauterization rat glaucoma model, Exp. Eye. Res, vol.82, pp.219-228, 2006.

S. Yu, T. Tanabe, and N. Yoshimura, A rat model of glaucoma induced by episcleral vein ligation, Exp. Eye. Res, vol.83, pp.758-770, 2006.

J. C. Morrison, C. Guo, W. O. Johnson, and E. C. , Pathophysiology of human glaucomatous optic nerve damage : insights from rodent models of glaucoma, Exp. Eye Res, vol.93, pp.156-64, 2011.

N. Nissirios, R. Chanis, J. E. Morrison, J. Cepurna, and W. O. , Comparison of anterior segment structures in two rat glaucoma models: an ultrasound biomicroscopic study, Invest. Ophthalmol. Vis. Sci, vol.49, pp.2478-2482, 2008.

A. S. Shifera, S. Trivedi, P. Chau, L. H. Bonnemaison, and R. Iguchi, Constitutive secretion of chemokines by cultured human trabecular meshwork cells, Exp. Eye Res, vol.91, pp.42-47, 2010.