Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2017

Open Access 01-12-2017 | Research

Microglial dynamics after axotomy-induced retinal ganglion cell death

Authors: Francisco M. Nadal-Nicolás, Manuel Jiménez-López, Manuel Salinas-Navarro, Paloma Sobrado-Calvo, Manuel Vidal-Sanz, Marta Agudo-Barriuso

Published in: Journal of Neuroinflammation | Issue 1/2017

Login to get access

Abstract

Background

Microglial cells (MCs) are the sentries of the central nervous system. In health, they are known as surveying MCs because they examine the tissue to maintain the homeostasis. In disease, they activate and, among other functions, become phagocytic to clean the cellular debris. In this work, we have studied the behavior of rat retinal MCs in two models of unilateral complete intraorbital optic nerve axotomy which elicit a different time course of retinal ganglion cell (RGC) loss.

Methods

Albino Sprague-Dawley rats were divided into these groups: (a) intact (no surgery), (b) fluorogold (FG) tracing from the superior colliculi, and (c) FG tracing + crush or transection of the left optic nerve. The retinas were dissected from 2 days to 2 months after the lesions (n = 4–12 group/lesion and time point) and then were subjected to Brn3a and Iba1 double immunodetection. In each intact retina, the total number of Brn3a+RGCs and Iba+MCs was quantified. In each traced retina (b and c groups), FG-traced RGCs and phagocytic microglial cells (PMCs, FG+Iba+) were also quantified. Topographical distribution was assessed by neighbor maps.

Results

In intact retinas, surveying MCs are homogenously distributed in the ganglion cell layer and the inner plexiform layer. Independently of the axotomy model, RGC death occurs in two phases, one quick and one protracted, and there is a lineal and topographical correlation between the appearance of PMCs and the loss of traced RGCs. Furthermore, the clearance of FG+RGCs by PMCs occurs 3 days after the actual loss of Brn3a expression that marks RGC death. In addition, almost 50% of MCs from the inner plexiform layer migrate to the ganglion cell layer during the quick phase of RGC loss, returning to the inner plexiform layer during the slow degeneration phase. Finally, in contrast to what happens in mice, in rats, there is no microglial phagocytosis in the contralateral uninjured retina.

Conclusions

Axotomy-induced RGC death occurs earlier than RGC clearance and there is an inverse correlation between RGC loss and PMC appearance, both numerically and topographically, suggesting that phagocytosis occurs as a direct response to RGC death rather than to axonal damage.
Literature
1.
go back to reference Vidal-Sanz M, Galindo-Romero C, Valiente-Soriano FJ, et al. Shared and differential retinal responses against optic nerve injury and ocular hypertension. Front Neurosci. 2017;11:235.CrossRefPubMedPubMedCentral Vidal-Sanz M, Galindo-Romero C, Valiente-Soriano FJ, et al. Shared and differential retinal responses against optic nerve injury and ocular hypertension. Front Neurosci. 2017;11:235.CrossRefPubMedPubMedCentral
2.
go back to reference Nadal-Nicolás FM, Sobrado-Calvo P, Jiménez-López M, et al. Long-term effect of optic nerve axotomy on the retinal ganglion cell layer. Invest Ophthalmol Vis Sci. 2015;56:6095–112.CrossRefPubMed Nadal-Nicolás FM, Sobrado-Calvo P, Jiménez-López M, et al. Long-term effect of optic nerve axotomy on the retinal ganglion cell layer. Invest Ophthalmol Vis Sci. 2015;56:6095–112.CrossRefPubMed
3.
go back to reference Villegas-Pérez MP, Vidal-Sanz M, Rasminsky M, et al. Rapid and protracted phases of retinal ganglion cell loss follow axotomy in the optic nerve of adult rats. J Neurobiol. 1993;24:23–36.CrossRefPubMed Villegas-Pérez MP, Vidal-Sanz M, Rasminsky M, et al. Rapid and protracted phases of retinal ganglion cell loss follow axotomy in the optic nerve of adult rats. J Neurobiol. 1993;24:23–36.CrossRefPubMed
4.
go back to reference Sánchez-Migallón MC, Valiente-Soriano FJ, Nadal-Nicolás FM, et al. Apoptotic retinal ganglion cell death after optic nerve transection or crush in mice: delayed RGC loss with BDNF or a caspase 3 inhibitor. Invest Ophthalmol Vis Sci. 2016;57:81–93.CrossRefPubMed Sánchez-Migallón MC, Valiente-Soriano FJ, Nadal-Nicolás FM, et al. Apoptotic retinal ganglion cell death after optic nerve transection or crush in mice: delayed RGC loss with BDNF or a caspase 3 inhibitor. Invest Ophthalmol Vis Sci. 2016;57:81–93.CrossRefPubMed
5.
go back to reference Berkelaar M, Clarke DB, Wang YC, et al. Axotomy results in delayed death and apoptosis of retinal ganglion cells in adult rats. J Neurosci. 1994;14:4368–74.PubMed Berkelaar M, Clarke DB, Wang YC, et al. Axotomy results in delayed death and apoptosis of retinal ganglion cells in adult rats. J Neurosci. 1994;14:4368–74.PubMed
6.
go back to reference Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26:239–57.CrossRefPubMedPubMedCentral Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26:239–57.CrossRefPubMedPubMedCentral
8.
go back to reference Nadal-Nicolás FM, Salinas-Navarro M, Jiménez-López M, et al. Displaced retinal ganglion cells in albino and pigmented rats. Front Neuroanat. 2014;6;8:99. Nadal-Nicolás FM, Salinas-Navarro M, Jiménez-López M, et al. Displaced retinal ganglion cells in albino and pigmented rats. Front Neuroanat. 2014;6;8:99.
9.
go back to reference Wang X, Zhao L, Zhang J, et al. Requirement for microglia for the maintenance of synaptic function and integrity in the mature retina. J Neurosci. 2016;36:2827–42.CrossRefPubMedPubMedCentral Wang X, Zhao L, Zhang J, et al. Requirement for microglia for the maintenance of synaptic function and integrity in the mature retina. J Neurosci. 2016;36:2827–42.CrossRefPubMedPubMedCentral
10.
go back to reference Wong WT. Microglial aging in the healthy CNS: phenotypes, drivers, and rejuvenation. Front Cell Neurosci. 2013;13(7):22. Wong WT. Microglial aging in the healthy CNS: phenotypes, drivers, and rejuvenation. Front Cell Neurosci. 2013;13(7):22.
11.
go back to reference Di Pierdomenico J, García-Ayuso D, Jiménez-López M, et al. Different ipsi- and contralateral glial responses to anti-VEGF and triamcinolone intravitreal injections in rats. Invest Ophthalmol Vis Sci. 2016;57:3533–44.CrossRefPubMed Di Pierdomenico J, García-Ayuso D, Jiménez-López M, et al. Different ipsi- and contralateral glial responses to anti-VEGF and triamcinolone intravitreal injections in rats. Invest Ophthalmol Vis Sci. 2016;57:3533–44.CrossRefPubMed
12.
go back to reference Mac Nair CE, Schlamp CL, Montgomery AD, et al. Retinal glial responses to optic nerve crush are attenuated in Bax-deficient mice and modulated by purinergic signaling pathways. J Neuroinflammation. 2016;13:93.CrossRefPubMedPubMedCentral Mac Nair CE, Schlamp CL, Montgomery AD, et al. Retinal glial responses to optic nerve crush are attenuated in Bax-deficient mice and modulated by purinergic signaling pathways. J Neuroinflammation. 2016;13:93.CrossRefPubMedPubMedCentral
13.
go back to reference Paques M, Simonutti M, Augustin S, et al. In vivo observation of the locomotion of microglial cells in the retina. Glia. 2010;58:1663–8.CrossRefPubMed Paques M, Simonutti M, Augustin S, et al. In vivo observation of the locomotion of microglial cells in the retina. Glia. 2010;58:1663–8.CrossRefPubMed
14.
go back to reference Chen L, Yang P, Kijlstra A. Distribution, markers, and functions of retinal microglia. Ocul Immunol Inflamm. 2002;10:27–39.CrossRefPubMed Chen L, Yang P, Kijlstra A. Distribution, markers, and functions of retinal microglia. Ocul Immunol Inflamm. 2002;10:27–39.CrossRefPubMed
15.
go back to reference Wolf SA, Boddeke HW, Kettenmann H. Microglia in physiology and disease. Annu Rev Physiol. 2017;79:619–43.CrossRefPubMed Wolf SA, Boddeke HW, Kettenmann H. Microglia in physiology and disease. Annu Rev Physiol. 2017;79:619–43.CrossRefPubMed
16.
go back to reference Colonna M, Butovsky O. Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol. 2017;35:441–68.CrossRefPubMed Colonna M, Butovsky O. Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol. 2017;35:441–68.CrossRefPubMed
17.
go back to reference Diaz-Aparicio I, Beccari S, Abiega O, Sierra A. Clearing the corpses: regulatory mechanisms, novel tools, and therapeutic potential of harnessing microglial phagocytosis in the diseased brain. Neural Regen Res. 2016;11:1533–9.CrossRefPubMedPubMedCentral Diaz-Aparicio I, Beccari S, Abiega O, Sierra A. Clearing the corpses: regulatory mechanisms, novel tools, and therapeutic potential of harnessing microglial phagocytosis in the diseased brain. Neural Regen Res. 2016;11:1533–9.CrossRefPubMedPubMedCentral
18.
go back to reference Zabel MK, Zhao L, Zhang Y, et al. Microglial phagocytosis and activation underlying photoreceptor degeneration is regulated by CX3CL1-CX3CR1 signaling in a mouse model of retinitis pigmentosa. Glia. 2016;64:1479–91.CrossRefPubMedPubMedCentral Zabel MK, Zhao L, Zhang Y, et al. Microglial phagocytosis and activation underlying photoreceptor degeneration is regulated by CX3CL1-CX3CR1 signaling in a mouse model of retinitis pigmentosa. Glia. 2016;64:1479–91.CrossRefPubMedPubMedCentral
19.
go back to reference Thanos S. The relationship of microglial cells to dying neurons during natural neuronal cell death and axotomy-induced degeneration of the rat retina. Eur J Neurosci. 1991;3:1189–207.CrossRefPubMed Thanos S. The relationship of microglial cells to dying neurons during natural neuronal cell death and axotomy-induced degeneration of the rat retina. Eur J Neurosci. 1991;3:1189–207.CrossRefPubMed
20.
go back to reference Thanos S, Pavlidis C, Mey J, et al. Specific transcellular staining of microglia in the adult rat after traumatic degeneration of carbocyanine-filled retinal ganglion cells. Exp Eye Res. 1992;55:101–17.CrossRefPubMed Thanos S, Pavlidis C, Mey J, et al. Specific transcellular staining of microglia in the adult rat after traumatic degeneration of carbocyanine-filled retinal ganglion cells. Exp Eye Res. 1992;55:101–17.CrossRefPubMed
21.
go back to reference Galindo-Romero C, Valiente-Soriano FJ, Jiménez-López M, et al. Effect of brain-derived neurotrophic factor on mouse axotomized retinal ganglion cells and phagocytic microglia. Invest Ophthalmol Vis Sci. 2013;54:974–85.CrossRefPubMed Galindo-Romero C, Valiente-Soriano FJ, Jiménez-López M, et al. Effect of brain-derived neurotrophic factor on mouse axotomized retinal ganglion cells and phagocytic microglia. Invest Ophthalmol Vis Sci. 2013;54:974–85.CrossRefPubMed
22.
23.
go back to reference Vidal-Sanz M, Nadal-Nicolas FM, Sobrado-Calvo P, et al. Quantitative and topographical microglial cell changes in the ganglion cell layer and inner plexiform layer after optic nerve axotomy. Invest Ophthalmol Vis Sci. 2016;57:2226. Vidal-Sanz M, Nadal-Nicolas FM, Sobrado-Calvo P, et al. Quantitative and topographical microglial cell changes in the ganglion cell layer and inner plexiform layer after optic nerve axotomy. Invest Ophthalmol Vis Sci. 2016;57:2226.
24.
go back to reference Nadal-Nicolás FM, Galindo-Romero C, Valiente-Soriano FJ, et al. Involvement of P2X7 receptor in neuronal degeneration triggered by traumatic injury. Sci Rep. 2016;6:38499.CrossRefPubMedPubMedCentral Nadal-Nicolás FM, Galindo-Romero C, Valiente-Soriano FJ, et al. Involvement of P2X7 receptor in neuronal degeneration triggered by traumatic injury. Sci Rep. 2016;6:38499.CrossRefPubMedPubMedCentral
25.
go back to reference Salinas-Navarro M, Mayor-Torroglosa S, Jiménez-López M, et al. A computerized analysis of the entire retinal ganglion cell population and its spatial distribution in adult rats. Vis Res. 2009;49:115–26.CrossRefPubMed Salinas-Navarro M, Mayor-Torroglosa S, Jiménez-López M, et al. A computerized analysis of the entire retinal ganglion cell population and its spatial distribution in adult rats. Vis Res. 2009;49:115–26.CrossRefPubMed
26.
go back to reference Nadal-Nicolás FM, Salinas-Navarro M, Vidal-Sanz M, et al. Two methods to trace retinal ganglion cells with fluorogold: from the intact optic nerve or by stereotactic injection into the optic tract. Exp Eye Res. 2015;131:12–9.CrossRefPubMed Nadal-Nicolás FM, Salinas-Navarro M, Vidal-Sanz M, et al. Two methods to trace retinal ganglion cells with fluorogold: from the intact optic nerve or by stereotactic injection into the optic tract. Exp Eye Res. 2015;131:12–9.CrossRefPubMed
27.
go back to reference Karlstetter M, Scholz R, Rutar M, et al. Retinal microglia: just bystander or target for therapy? Prog Retin Eye Res. 2015;45:30–57.CrossRefPubMed Karlstetter M, Scholz R, Rutar M, et al. Retinal microglia: just bystander or target for therapy? Prog Retin Eye Res. 2015;45:30–57.CrossRefPubMed
28.
29.
30.
go back to reference Ellis-Behnke RG, Jonas RA, Jonas JB. The microglial system in the eye and brain in response to stimuli in vivo. J Glaucoma. 2013;22(Suppl 5):S32–5.CrossRefPubMed Ellis-Behnke RG, Jonas RA, Jonas JB. The microglial system in the eye and brain in response to stimuli in vivo. J Glaucoma. 2013;22(Suppl 5):S32–5.CrossRefPubMed
31.
go back to reference Marín-Teva JL, Cuadros MA, Martín-Oliva D, et al. Microglia and neuronal cell death. Neuron Glia Biol. 2011;7:25–40.CrossRefPubMed Marín-Teva JL, Cuadros MA, Martín-Oliva D, et al. Microglia and neuronal cell death. Neuron Glia Biol. 2011;7:25–40.CrossRefPubMed
32.
go back to reference Ramirez AI, de Hoz R, Salobrar-Garcia E, et al. The role of microglia in retinal neurodegeneration: Alzheimer’s disease, Parkinson, and glaucoma. Front Aging Neurosci. 2017; 10.3389/fnagi.2017.00214. Ramirez AI, de Hoz R, Salobrar-Garcia E, et al. The role of microglia in retinal neurodegeneration: Alzheimer’s disease, Parkinson, and glaucoma. Front Aging Neurosci. 2017; 10.​3389/​fnagi.​2017.​00214.
33.
go back to reference Baptista FI, Aveleira CA, Castilho ÁF, et al. Elevated glucose and interleukin-1β differentially affect retinal microglial cell proliferation. Mediat Inflamm. 2017;4316316:2017. Baptista FI, Aveleira CA, Castilho ÁF, et al. Elevated glucose and interleukin-1β differentially affect retinal microglial cell proliferation. Mediat Inflamm. 2017;4316316:2017.
34.
go back to reference Nadal-Nicolás FM, Jiménez-López M, Sobrado-Calvo P, et al. Brn3a as a marker of retinal ganglion cells: qualitative and quantitative time course studies in naive and optic nerve-injured retinas. Invest Ophthalmol Vis Sci. 2009;50:3860–8.CrossRefPubMed Nadal-Nicolás FM, Jiménez-López M, Sobrado-Calvo P, et al. Brn3a as a marker of retinal ganglion cells: qualitative and quantitative time course studies in naive and optic nerve-injured retinas. Invest Ophthalmol Vis Sci. 2009;50:3860–8.CrossRefPubMed
35.
go back to reference Salinas-Navarro M, Jiménez-López M, Valiente-Soriano FJ, et al. Retinal ganglion cell population in adult albino and pigmented mice: a computerized analysis of the entire population and its spatial distribution. Vis Res. 2009;49:637–47.CrossRefPubMed Salinas-Navarro M, Jiménez-López M, Valiente-Soriano FJ, et al. Retinal ganglion cell population in adult albino and pigmented mice: a computerized analysis of the entire population and its spatial distribution. Vis Res. 2009;49:637–47.CrossRefPubMed
36.
go back to reference Parrilla-Reverter G, Agudo M, Nadal-Nicolás F, et al. Time-course of the retinal nerve fibre layer degeneration after complete intra-orbital optic nerve transection or crush: a comparative study. Vis Res. 2009;49:2808–25.CrossRefPubMed Parrilla-Reverter G, Agudo M, Nadal-Nicolás F, et al. Time-course of the retinal nerve fibre layer degeneration after complete intra-orbital optic nerve transection or crush: a comparative study. Vis Res. 2009;49:2808–25.CrossRefPubMed
37.
go back to reference Nadal-Nicolás FM, Jiménez-López M, Salinas-Navarro M, et al. Whole number, distribution and co-expression of brn3 transcription factors in retinal ganglion cells of adult albino and pigmented rats. PLoS One. 2012;7(11):e49830.CrossRefPubMedPubMedCentral Nadal-Nicolás FM, Jiménez-López M, Salinas-Navarro M, et al. Whole number, distribution and co-expression of brn3 transcription factors in retinal ganglion cells of adult albino and pigmented rats. PLoS One. 2012;7(11):e49830.CrossRefPubMedPubMedCentral
38.
go back to reference Nadal-Nicolás FM, Rodriguez-Villagra E, Bravo-Osuna I, et al. Ketorolac administration attenuates retinal ganglion cell death after axonal injury. Invest Ophthalmol Vis Sci. 2016;57:1183–92.CrossRefPubMed Nadal-Nicolás FM, Rodriguez-Villagra E, Bravo-Osuna I, et al. Ketorolac administration attenuates retinal ganglion cell death after axonal injury. Invest Ophthalmol Vis Sci. 2016;57:1183–92.CrossRefPubMed
39.
go back to reference Nadal-Nicolás FM, Madeira MH, Salinas-Navarro M, et al. Transient downregulation of melanopsin expression after retrograde tracing or optic nerve injury in adult rats. Invest Ophthalmol Vis Sci. 2015;56:4309–23.CrossRefPubMed Nadal-Nicolás FM, Madeira MH, Salinas-Navarro M, et al. Transient downregulation of melanopsin expression after retrograde tracing or optic nerve injury in adult rats. Invest Ophthalmol Vis Sci. 2015;56:4309–23.CrossRefPubMed
41.
go back to reference Gómez-Ramírez AM, Villegas-Pérez MP, Miralles de Imperial J, et al. Effects of intramuscular injection of botulinum toxin and doxorubicin on the survival of abducens motoneurons. Invest Ophthalmol Vis Sci. 1999;40:414–24.PubMed Gómez-Ramírez AM, Villegas-Pérez MP, Miralles de Imperial J, et al. Effects of intramuscular injection of botulinum toxin and doxorubicin on the survival of abducens motoneurons. Invest Ophthalmol Vis Sci. 1999;40:414–24.PubMed
42.
go back to reference Sellés-Navarro I, Villegas-Pérez MP, Salvador-Silva M, et al. Retinal ganglion cell death after different transient periods of pressure-induced ischemia and survival intervals. A quantitative in vivo study. Invest Ophthalmol Vis Sci. 1996;37:2002–14.PubMed Sellés-Navarro I, Villegas-Pérez MP, Salvador-Silva M, et al. Retinal ganglion cell death after different transient periods of pressure-induced ischemia and survival intervals. A quantitative in vivo study. Invest Ophthalmol Vis Sci. 1996;37:2002–14.PubMed
43.
go back to reference Ortín-Martínez A, Salinas-Navarro M, Nadal-Nicolás FM, et al. Laser-induced ocular hypertension in adult rats does not affect non-RGC neurons in the ganglion cell layer but results in protracted severe loss of cone-photoreceptors. Exp Eye Res. 2015;132:17–33.CrossRefPubMed Ortín-Martínez A, Salinas-Navarro M, Nadal-Nicolás FM, et al. Laser-induced ocular hypertension in adult rats does not affect non-RGC neurons in the ganglion cell layer but results in protracted severe loss of cone-photoreceptors. Exp Eye Res. 2015;132:17–33.CrossRefPubMed
44.
go back to reference Janssen KT, Mac Nair CE, Dietz JA, et al. Nuclear atrophy of retinal ganglion cells precedes the bax-dependent stage of apoptosis. Invest Ophthalmol Vis Sci. 2013;54:1805–15.CrossRefPubMedPubMedCentral Janssen KT, Mac Nair CE, Dietz JA, et al. Nuclear atrophy of retinal ganglion cells precedes the bax-dependent stage of apoptosis. Invest Ophthalmol Vis Sci. 2013;54:1805–15.CrossRefPubMedPubMedCentral
45.
go back to reference He T, Mortensen X, Wang P, et al. The effects of immune protein CD3ζ development and degeneration of retinal neurons after optic nerve injury. PLoS One. 2017;12(4):e0175522.CrossRefPubMedPubMedCentral He T, Mortensen X, Wang P, et al. The effects of immune protein CD3ζ development and degeneration of retinal neurons after optic nerve injury. PLoS One. 2017;12(4):e0175522.CrossRefPubMedPubMedCentral
46.
go back to reference Sobrado-Calvo P, Vidal-Sanz M, Villegas-Pérez MP. Rat retinal microglial cells under normal conditions, after optic nerve section, and after optic nerve section and intravitreal injection of trophic factors or macrophage inhibitory factor. J Comp Neurol. 2007;501(6):866–78.CrossRefPubMed Sobrado-Calvo P, Vidal-Sanz M, Villegas-Pérez MP. Rat retinal microglial cells under normal conditions, after optic nerve section, and after optic nerve section and intravitreal injection of trophic factors or macrophage inhibitory factor. J Comp Neurol. 2007;501(6):866–78.CrossRefPubMed
47.
go back to reference Cen LP, Han M, Zhou L, et al. Bilateral retinal microglial response to unilateral optic nerve transection in rats. Neuroscience. 2015;311:56–66.CrossRefPubMed Cen LP, Han M, Zhou L, et al. Bilateral retinal microglial response to unilateral optic nerve transection in rats. Neuroscience. 2015;311:56–66.CrossRefPubMed
48.
go back to reference Garcia-Valenzuela E, Sharma SC, Piña AL. Multilayered retinal microglial response to optic nerve transection in rats. Mol Vis. 2005;11:225–31.PubMed Garcia-Valenzuela E, Sharma SC, Piña AL. Multilayered retinal microglial response to optic nerve transection in rats. Mol Vis. 2005;11:225–31.PubMed
49.
go back to reference Santos AM, Calvente R, Tassi M, et al. Embryonic and postnatal development of microglial cells in the mouse retina. J Comp Neurol. 2008;506:224–39.CrossRefPubMed Santos AM, Calvente R, Tassi M, et al. Embryonic and postnatal development of microglial cells in the mouse retina. J Comp Neurol. 2008;506:224–39.CrossRefPubMed
50.
go back to reference Lee JE, Liang KJ, Fariss RN, et al. Ex vivo dynamic imaging of retinal microglia using time-lapse confocal microscopy. Invest Ophthalmol Vis Sci. 2008;49:4169–76.CrossRefPubMedPubMedCentral Lee JE, Liang KJ, Fariss RN, et al. Ex vivo dynamic imaging of retinal microglia using time-lapse confocal microscopy. Invest Ophthalmol Vis Sci. 2008;49:4169–76.CrossRefPubMedPubMedCentral
51.
go back to reference Bodeutsch N, Siebert H, Dermon C, et al. Unilateral injury to the adult rat optic nerve causes multiple cellular responses in the contralateral site. J Neurobiol. 1999;38:116–28.CrossRefPubMed Bodeutsch N, Siebert H, Dermon C, et al. Unilateral injury to the adult rat optic nerve causes multiple cellular responses in the contralateral site. J Neurobiol. 1999;38:116–28.CrossRefPubMed
52.
go back to reference Ito D, Imai Y, Ohsawa K, et al. Microglia-specific localisation of a novel calcium binding protein, Iba1. Brain Res Mol Brain Res. 1998;57:1–9.CrossRefPubMed Ito D, Imai Y, Ohsawa K, et al. Microglia-specific localisation of a novel calcium binding protein, Iba1. Brain Res Mol Brain Res. 1998;57:1–9.CrossRefPubMed
53.
go back to reference Panagis L, Thanos S, Fischer D, et al. Unilateral optic nerve crush induces bilateral retinal glial cell proliferation. Eur J Neurosci. 2005;21:2305–9.CrossRefPubMed Panagis L, Thanos S, Fischer D, et al. Unilateral optic nerve crush induces bilateral retinal glial cell proliferation. Eur J Neurosci. 2005;21:2305–9.CrossRefPubMed
54.
go back to reference Liu S, Li ZW, Weinreb RN, et al. Tracking retinal microgliosis in models of retinal ganglion cell damage. Invest Ophthalmol Vis Sci. 2012;53:6254–62.CrossRefPubMed Liu S, Li ZW, Weinreb RN, et al. Tracking retinal microgliosis in models of retinal ganglion cell damage. Invest Ophthalmol Vis Sci. 2012;53:6254–62.CrossRefPubMed
55.
go back to reference Ramírez AI, Salazar JJ, de Hoz R, et al. Quantification of the effect of different levels of IOP in the astroglia of the rat retina ipsilateral and contralateral to experimental glaucoma. Invest Ophthalmol Vis Sci. 2010;51:5690–6.CrossRefPubMed Ramírez AI, Salazar JJ, de Hoz R, et al. Quantification of the effect of different levels of IOP in the astroglia of the rat retina ipsilateral and contralateral to experimental glaucoma. Invest Ophthalmol Vis Sci. 2010;51:5690–6.CrossRefPubMed
56.
go back to reference Müller M, Holländer H. A small population of retinal ganglion cells projecting to the retina of the other eye. An experimental study in the rat and the rabbit. Exp Brain Res. 1988;71:611–7.CrossRefPubMed Müller M, Holländer H. A small population of retinal ganglion cells projecting to the retina of the other eye. An experimental study in the rat and the rabbit. Exp Brain Res. 1988;71:611–7.CrossRefPubMed
57.
go back to reference Avellaneda-Chevrier VK, Wang X, Hooper ML, et al. The retino-retinal projection: tracing retinal ganglion cells projecting to the contralateral retina. Neurosci Lett. 2015;591:105–9.CrossRefPubMed Avellaneda-Chevrier VK, Wang X, Hooper ML, et al. The retino-retinal projection: tracing retinal ganglion cells projecting to the contralateral retina. Neurosci Lett. 2015;591:105–9.CrossRefPubMed
58.
go back to reference Nadal-Nicolás FM, Valiente-Soriano FJ, Salinas-Navarro M, et al. Retino-retinal projection in juvenile and young adult rats and mice. Exp Eye Res. 2015;134:47–52.CrossRefPubMed Nadal-Nicolás FM, Valiente-Soriano FJ, Salinas-Navarro M, et al. Retino-retinal projection in juvenile and young adult rats and mice. Exp Eye Res. 2015;134:47–52.CrossRefPubMed
59.
go back to reference Thanos S, Richter W. The migratory potential of vitally labelled microglial cells within the retina of rats with hereditary photoreceptor dystrophy. Int J Dev Neurosci. 1993;11:671–80.CrossRefPubMed Thanos S, Richter W. The migratory potential of vitally labelled microglial cells within the retina of rats with hereditary photoreceptor dystrophy. Int J Dev Neurosci. 1993;11:671–80.CrossRefPubMed
60.
go back to reference Madeira MH, Boia R, Santos PF, et al. Contribution of microglia-mediated neuroinflammation to retinal degenerative diseases. Mediat Inflamm. 2015;673090:2015. Madeira MH, Boia R, Santos PF, et al. Contribution of microglia-mediated neuroinflammation to retinal degenerative diseases. Mediat Inflamm. 2015;673090:2015.
61.
go back to reference Chen Z, Jalabi W, Shpargel KB, et al. Lipopolysaccharide-induced microglial activation and neuroprotection against experimental brain injury is independent of hematogenous TLR4. J Neurosci. 2012;32:11706–15.CrossRefPubMedPubMedCentral Chen Z, Jalabi W, Shpargel KB, et al. Lipopolysaccharide-induced microglial activation and neuroprotection against experimental brain injury is independent of hematogenous TLR4. J Neurosci. 2012;32:11706–15.CrossRefPubMedPubMedCentral
62.
go back to reference Karlstetter M, Ebert S, Langmann T. Microglia in the healthy and degenerating retina: insights from novel mouse models. Immunobiology. 2010;215:685–91.CrossRefPubMed Karlstetter M, Ebert S, Langmann T. Microglia in the healthy and degenerating retina: insights from novel mouse models. Immunobiology. 2010;215:685–91.CrossRefPubMed
63.
go back to reference Madeira MH, Elvas F, Boia R, et al. Adenosine A2AR blockade prevents neuroinflammation-induced death of retinal ganglion cells caused by elevated pressure. J Neuroinflammation. 2015;12:115.CrossRefPubMedPubMedCentral Madeira MH, Elvas F, Boia R, et al. Adenosine A2AR blockade prevents neuroinflammation-induced death of retinal ganglion cells caused by elevated pressure. J Neuroinflammation. 2015;12:115.CrossRefPubMedPubMedCentral
64.
go back to reference Davis BM, Salinas-Navarro M, Cordeiro MF, et al. Characterizing microglia activation: a spatial statistics approach to maximize information extraction. Sci Rep. 2017;7:1576.CrossRefPubMedPubMedCentral Davis BM, Salinas-Navarro M, Cordeiro MF, et al. Characterizing microglia activation: a spatial statistics approach to maximize information extraction. Sci Rep. 2017;7:1576.CrossRefPubMedPubMedCentral
65.
go back to reference Jonas RA, Yuan TF, Liang YX, et al. The spider effect: morphological and orienting classification of microglia in response to stimuli in vivo. PLoS One. 2012;7(2):e30763.CrossRefPubMedPubMedCentral Jonas RA, Yuan TF, Liang YX, et al. The spider effect: morphological and orienting classification of microglia in response to stimuli in vivo. PLoS One. 2012;7(2):e30763.CrossRefPubMedPubMedCentral
66.
go back to reference de Hoz R, Gallego BI, Ramírez AI, et al. Rod-like microglia are restricted to eyes with laser-induced ocular hypertension but absent from the microglial changes in the contralateral untreated eye. PLoS One. 2013;8(12):e83733.CrossRefPubMedPubMedCentral de Hoz R, Gallego BI, Ramírez AI, et al. Rod-like microglia are restricted to eyes with laser-induced ocular hypertension but absent from the microglial changes in the contralateral untreated eye. PLoS One. 2013;8(12):e83733.CrossRefPubMedPubMedCentral
67.
go back to reference Bachstetter AD, Ighodaro ET, Hassoun Y, et al. Rod-shaped microglia morphology is associated with aging in 2 human autopsy series. Neurobiol Aging. 2017;52:98–105.CrossRefPubMed Bachstetter AD, Ighodaro ET, Hassoun Y, et al. Rod-shaped microglia morphology is associated with aging in 2 human autopsy series. Neurobiol Aging. 2017;52:98–105.CrossRefPubMed
68.
go back to reference Trapp BD, Wujek JR, Criste GA, et al. Evidence for synaptic stripping by cortical microglia. Glia. 2007;55:360–8.CrossRefPubMed Trapp BD, Wujek JR, Criste GA, et al. Evidence for synaptic stripping by cortical microglia. Glia. 2007;55:360–8.CrossRefPubMed
69.
go back to reference Garcia-Valenzuela E, Sharma SC. Laminar restriction of retinal macrophagic response to optic nerve axotomy in the rat. J Neurobiol. 1999;40:55–66.CrossRefPubMed Garcia-Valenzuela E, Sharma SC. Laminar restriction of retinal macrophagic response to optic nerve axotomy in the rat. J Neurobiol. 1999;40:55–66.CrossRefPubMed
70.
go back to reference Wohl SG, Schmeer CW, Witte OW, et al. Proliferative response of microglia and macrophages in the adult mouse eye after optic nerve lesion. Invest Ophthalmol Vis Sci. 2010;51:2686–96.CrossRefPubMed Wohl SG, Schmeer CW, Witte OW, et al. Proliferative response of microglia and macrophages in the adult mouse eye after optic nerve lesion. Invest Ophthalmol Vis Sci. 2010;51:2686–96.CrossRefPubMed
71.
go back to reference Wohl SG, Schmeer CW, Friese T, et al. In situ dividing and phagocytosing retinal microglia express nestin, vimentin, and NG2 in vivo. PLoS One. 2011;6(8):e22408.CrossRefPubMedPubMedCentral Wohl SG, Schmeer CW, Friese T, et al. In situ dividing and phagocytosing retinal microglia express nestin, vimentin, and NG2 in vivo. PLoS One. 2011;6(8):e22408.CrossRefPubMedPubMedCentral
72.
go back to reference Trost A, Motloch K, Bruckner D, et al. Time-dependent retinal ganglion cell loss, microglial activation and blood-retina-barrier tightness in an acute model of ocular hypertension. Exp Eye Res. 2015;136:59–71.CrossRefPubMed Trost A, Motloch K, Bruckner D, et al. Time-dependent retinal ganglion cell loss, microglial activation and blood-retina-barrier tightness in an acute model of ocular hypertension. Exp Eye Res. 2015;136:59–71.CrossRefPubMed
73.
go back to reference Zhang Y, Zhao L, Wang X, et al. Microglia homeostasis in the adult mouse retina: restoration of microglial distribution, morphology, and function following acute depletion. Invest Ophthalmol Vis Sci. 2017;58:806. Zhang Y, Zhao L, Wang X, et al. Microglia homeostasis in the adult mouse retina: restoration of microglial distribution, morphology, and function following acute depletion. Invest Ophthalmol Vis Sci. 2017;58:806.
74.
go back to reference Hilla AM, Diekmann H, Fischer D. Microglia are irrelevant for neuronal degeneration and axon regeneration after acute injury. J Neurosci. 2017;37:6113–24.CrossRefPubMed Hilla AM, Diekmann H, Fischer D. Microglia are irrelevant for neuronal degeneration and axon regeneration after acute injury. J Neurosci. 2017;37:6113–24.CrossRefPubMed
Metadata
Title
Microglial dynamics after axotomy-induced retinal ganglion cell death
Authors
Francisco M. Nadal-Nicolás
Manuel Jiménez-López
Manuel Salinas-Navarro
Paloma Sobrado-Calvo
Manuel Vidal-Sanz
Marta Agudo-Barriuso
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2017
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-017-0982-7

Other articles of this Issue 1/2017

Journal of Neuroinflammation 1/2017 Go to the issue