* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download View PDF
Psychoneuroimmunology wikipedia , lookup
Development of the nervous system wikipedia , lookup
Axon guidance wikipedia , lookup
Synaptogenesis wikipedia , lookup
Subventricular zone wikipedia , lookup
Signal transduction wikipedia , lookup
Clinical neurochemistry wikipedia , lookup
Feature detection (nervous system) wikipedia , lookup
Neuropsychopharmacology wikipedia , lookup
Neuroprotection in glaucoma: present and future CHEN Shi-da1, WANG Lu1, and ZHANG Xiu-lan1* 1 State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, P.R. China; * Corresponding to: Xiulan Zhang, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54S. Xian lie Road, Guangzhou 510060, P.R.China; [email protected]. Tel:8620--87330484; Fax:8620-87333271. Acknowledgments: This research was supported by the grants from the National Natural Science Foundation of China (81170849) and the Fundamental Research Funds of State Key Laboratory of Ophthalmology (2011C02). Keywords: glaucoma; neuroprotection; retinal ganglion cells Abstract Objective: To review the updated research on neuroprotection in glaucoma, and summarize the potential agents investigated so far. Data sources: The data in this review were collected from PubMed and Google Scholar databases published in English up to September 2012, with key words including glaucoma, neuroprotection, and retinal ganglion cell (RGC), both alone and in combination. Publications from the past ten years were selected, but important older articles were not excluded. Study selection: Articles about neuroprotection in glaucoma were selected and reviewed, and reference lists of articles identified by this search strategy and judged relevant to this review were also included. Results: Although lowering the intraocular pressure is the only therapy approved as being effective in the treatment of glaucoma, increasing numbers of studies have discovered various mechanisms of RGC death in the glaucoma and relevant neuroprotective strategies. These strategies target neurotrophic factor deprivation, excitotoxic damage, oxidative stress, mitochondrial dysfunction, inflammation, activation of intrinsic and extrinsic apoptotic signals, ischemia and protein misfolding. Exploring the mechanism of axonal transport failure, synaptic dysfunction, the glial system in glaucoma, and stem cell used in glaucoma constitute promising research areas of the future. Conclusions: Neuroprotective strategies continue to be refined, and future deep investment in researching the pathogenesis of glaucoma may provide novel and practical neuroprotection tactics. Establishing a system to assess the effects of neuroprotection treatments may further facilitate this research. INTRODUCTION Glaucoma is the second leading cause of blindness worldwide1. It is recognized to be an optic neuropathy accompanied by typical structural and functional defects (optic disc damage and visual field loss) in at least one eye2. It is widely accepted that the death of a substantial number of retinal ganglion cell (RGCs) in the inner retina and the loss of their axons in the optic nerve are the pathophysiologic characteristics of glaucoma2. Several well-known clinical studies have demonstrated that intraocular pressure (IOP) lowering is currently the only effective treatment of primary open-angle glaucoma (POAG) and of ocular hypertension; the treatment not only delays or prevents the onset of POAG in individuals with elevated IOP, but also partly prevents visual loss in cases of POAG. Nonetheless, the reduction of IOP is not always successful and many patients continue to experience progressive visual loss, as indicated in the OHTS (Ocular Hypertension Treatment Study), CIGTS (Collaborative Initial Glaucoma Treatment Study), and EMGT (Early Manifest Glaucoma Trial)3,4,5,6. Moreover, achieving adequate pressure lowering may be difficult or may be associated with adverse effects7. Neuroprotection offers potential as a complementary therapy to IOP lowering, and also serves as an alternative therapy for when IOP-lowering agents are not used, not tolerated, or not effective8. Generally, IOP lowering could obstruct or delay the progress of visual loss in the glaucoma, and it is indirectly neuroprotective. However, by definition, glaucoma neuroprotection is considered to be independent of IOP lowering, as it directly targets neurons within the central visual pathway, especially the RGCs8,9. Neuroprotective agents focus on the multiple pathogenic mechanisms that result in axonal degeneration and RGC death. Mechanisms of RGC death in glaucoma and neuroprotection strategies The molecular basis of RGC death in the glaucoma stemming from animal models include neurotrophic factor deprivation, excitotoxic damage, oxidative stress, mitochondrial dysfunction, inflammation, activation of intrinsic and extrinsic apoptotic signals, ischemia, protein misfolding, axonal transport failure, synaptic dysfunction, and misbehaved glial system in retina. There has been considerable progress in our understanding of the multiple pathways that lead to RGC death in glaucoma. 1. Neurotrophins Neurotrophins play a key role in the development, differentiation, and survival of RGCs; they include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin3/4/5 (NT-3/4/5). The biological effects of these neurotrophins are mediated by two classes of cell surface receptors: 1) the tropomyosin-related kinase (Trk) family, consisting of TrkA, the receptor for NGF, TrkB, the receptor for BDNF and NT-4/5, and TrkC, the receptor for NT-3; and 2) the p75 receptor (p75NTR), which binds all neurotrophins with a similar affinity10. Activation of Trk receptors has been typically associated with cell survival, while p75NTR can stimulate both survival and apoptotic pathways11,12. Anything that induced the neurotrophins deprivation—including interrupted transportation from the upper neurons, low expression level of receptor, and gene mutation—will cause RGC death, and thus delivering neurotrophins to the RGCs will have a protective effect. NTF4 gene mutations have been found in 1.7% of POAG patients of European origin; the mutations showed a decreased affinity of NT-4 with TrkB, thus leading to disease progression13. One study finds that after receiving murine neurotrophic growth factor eyedrops once daily for three months, three advanced glaucoma patients showed improvement in all parameters of visual function14. Among the neurotrophins, BDNF has received particular attention because of its potential role in the survival of RGCs. BDNF promotes neuronal survival by inhibiting default apoptotic pathways. The direct application of recombinant the BDNF or viral vector-mediated BDNF expression has been found to protect RGCs in the optic nerve section model or high IOP glaucoma model15,16. Glial cell line-derived neurotrophic factor (GDNF), another neurotrophin, is less effective than BDNF in protecting injured RGCs, but when GDNF is combined with BDNF, the effect is almost doubled compared to the independent administration of each neurotrophic factor17. BDNF is also an important regulator of synaptic plasticity18, and of position-dependent branching of developing RGC axons19. In the retina, BDNF contributes to the shaping and maintenance of RGC dendritic morphology following axonal injury20. Ciliary neurotrophic factor (CNTF) is a cytoplasmic protein that acts as an endogenous response to retinal neurons under pathological conditions. In the adult rat retina, CNTF is primarily localized in Müller cells, and its expression increased after axotomy and ischemia, and in experimental glaucoma21,22. Overexpression of CNTF promotes RGC survival in laser-induced glaucoma models23. Exogenous CNTF exhibits moderate protection for RGCs in different models23,24; however, unlike BDNF, CNTF is able to stimulate RGC axon regeneration after injury25. One disappointing fact is that excess exogenous CNTF could impair visual function; thus the concentration of CNTF used in the retina must be monitored26. However, there are obstacles to using the neurotrophins in the eyes, including 1) the short half-time of these neurotrophins; 2) the low permeability of the blood-brain barrier; 3) the demand for high doses; 4) the insufficient to stimulate the regrowth of injured RGC axons; and 5) the low level of the neurotrophin receptor in injured RGCs. As a result, most of the research currently focuses on 1) developing a novel drug delivery, in the hope of increasing the drug concentration for the eye and raising the function time; 2) finding a new neurotrophic analogue, such as the small peptide, which has a high facility to cross the blood-brain barrier and a longer half-time27,28(the small peptides ADNF-9 and NAP not only increased the survival but also supported neurite outgrowth of rat RGCs in vitro29); 3) facilitating the selective upregulation of TrkB in RGCs combined with exogenous or endogenous BDNF to enhance the duration of level of BDNF-induced neuroprotection30; and 4) combining the application of factors with independent mechanisms, which has shown that the combination of basic fibroblast growth factor, neurotrophin-3, and BDGF delivered to the RGC not only prevents RGC loss but also promotes axon growth after CNS injury. This effect exceeds that of the each neurotrophic factor alone31. 2. Excitotoxicity It is universally accepted that abnormal high extracellular levels of glutamate could activate the NMDA receptor (NMDAR ) channels in RGCs, leading to the deleterious increase of intracellular calcium and nitric oxide production, and the activation of pro-apoptotic signaling cascades, which result in the death of RGCs32. A great number of studies have demonstrated that exogenous NMDA induced the rapid death of adult RGCs, and excess glutamate has been found in the retina in glaucoma33, while inhibitors of NMDAR or downstream pathways have a protective role in experimental models of retinal ischemia and glaucoma34, 35. However, several studies demonstrated that there was no excessive glutamate found in experimental animals with ocular hypertension or in human glaucoma36,37. Thus, there may be no drastic elevation or accumulation of glutamate in chronic and progressive glaucoma, and a glutamate increase is likely to occur in the localized area of the retina at any one time. On the other hand, extracellular Mg2+ regulates the NMDAR under different membrane potential, which may lead to NMDAR activation even at physiological levels of glutamate38, 39. This contradiction reminds us of how complex a mechanism glutamate is to the RGCs, and how it is not enough to simply rule out an excitotoxic component in RGCs to achieve a protective effect. The glutamate will not only be released by astrocyte or microglial cells exposed to ischemia or high IOP, but the initial degeneration of some RGCs seems to lead to a toxic environment such as glutamate excitotoxicity32. This may be the reason that injured RGCs result in secondary damage to the surrounding, normal RGCs, which in turn leading to progress of glaucoma. Modulation of the NMDA receptor has always been a major area of research in glaucoma neuroprotection. There are several anti-excitotoxic drugs that have been investigated to have a neuroprotective effect. Memantine, an uncompetitive antagonist of the NMDA receptor that was approved by the FDA for treating Alzheimer’s disease, blocks only excessive NMDA receptor activity while displaying weak potency during normal synaptic transmission by glutamate9,33. It had been found to protect against optic nerve fiber loss, neuronal shrinkage within the central visual pathway, and loss of visual function in monkey hypertension glaucoma models40, 41. Nevertheless, the result of a phase Ⅲ clinical trial in the human open-angle glaucoma was disappointing, which may be due to the inappropriate endpoint and insufficient duration of the study42.Another non-competitive antagonist of the NMDA receptor is MK801, which shows a strong protective effect on the RGCs both in vitro and in vivo43, 44. However, because of its high affinity with the NMDA receptors and long dwell time in the channel, which results in a large neurotoxic effect, it cannot be used clinically. Finding a drug with the same protective effect as MK801 but with less neurotoxic effect or combining MK801 with other agents to reduce the adverse effect would be helpful. 3. Oxidative stress Oxidative stress, characterized by the imbalance between the production of reactive oxygen species (ROS) and their elimination system, plays an essential role in the injury and death of neuron cells, including RGCs. Excessive ROS could cause protein modification and DNA damage, consequently activating cell death signals45. ROS can be caused by mitochondrial dysfunction, abnormal protein folding, and a defective ubiquitination and proteasome degradation system46. It has been confirmed that oxidative damage occurs in experimental models of optic nerve injury and in human glaucoma, and insufficiency in ROS-neutralizing mechanisms in RGCs were also discovered in glaucoma47-49. As a result, keeping the ROS production system and clear-up system balance would benefit the RGCs’ survival50, 51. The regulation of cellular redox status is provided by the glutathione and the thioredoxin (TRX) systems. Overexpression of TRX1 and TRX2 protected RGCs from pharmacologically-induced oxidative stress, ocular hypertension52. Melatonin, a potent, naturally occurring antioxidant with free-radical scavenging activity, displays a critical role in aqueous humor circulation and shows potential as a neuroprotectant. Melatonin was shown to prevent RGC loss in the NO-induced retinal damage model53. Ginkgo Biloba, which is part of traditional Chinese medicine, has been used to treat Alzheimer’s disease and low-tension glaucoma54, 55. Its extract EGb761 showed robust antioxidant qualities, inhibiting chemically induced apoptosis. Moreover, EGb761 decreased high IOP-induced RGC loss56, 57. 4. Mitochondrial dysfunction The mitochondrial is an organelle in which the interaction between anti- and pro-apoptotic Bcl-2 family members occurs. When the trend is toward apoptosis, mitochondrial membrane permeability increases and releases cytochrome c, subsequently forming an apoptosome with Apaf-1 and procaspase-9 and leading to cell death. The early release of cytochrome c in injured RGCs was showed in optic nerve axotomy58. However, the specific role of mitochondrial mediators in RGC death in glaucoma remains poorly explored. RGCs have a high metabolic activity and energy demand, which is confirmed by the large amount of mitochondrial in RGC soma and the intraretinal portion of their axons59. Mitochondrial DNA damage increases with age and with the reduced production of ATP in the RGC, compromising RGC viability. As a result, mitochondrial dysfunction could play a key role in glaucoma. It is found that decreased ATP availability leads to cellular energy crisis, interrupting the normal transduction of action potential along RGC axons60. ATP content in the mouse optic nerve dropped with age and high IOP in DBA/2J mice, leading to RGC axon dysfunction61. However, excess ATP release from injured and dying cells to the extracellular place, thus activating the 2X7 receptor, would be toxic to the RGCs62. Mitochondrial biogenesis-based neuroprotection is intended to restore the balance of ATP levels, increasing their available energy supply63, 64. PGC-1α is a key mitochondrial biogenesis factor. The regulation of PGC-1α has been explored in many mitochondrial-related diseases, such as cancer, neurodegenerative diseases, cardiovascular disease65, it remains poorly explored in glaucoma64. Coenzyme Q10, a component of the mitochondrial electron transport chain that promotes ATP production, is multifactorial in its neuroprotection of RGCs. It has been confirmed that Coenzyme Q10 not only mediates the electron transport chain but also displays antioxidant properties, which show a strong protective effect for RGCs both in vivo and in vitro66, 67. 5. Inflammation and immunological strategies In the CNS, boosting a self-specific immune response promotes recovery. Growing evidence in clinical and experimental studies strongly suggest the involvement of the immune system in glaucoma68. Present research findings collectively suggest that innate immune cells, autoreactive T cells, autoantibodies, and complement activation may impair RGCs and increase the susceptibility to disease progression in glaucoma68, 69. And the role of glial cell in the inflammation and immune response in the glaucoma would discuss in another section bellow. TNF-α is a pro-inflammatory cytokine that acts on two distinct receptors, TNFR1 and TNFR2. It serves as a critical element in immune homeostasis and in mediating apoptosis. In the retina, TNF-α is mainly produced by Müller glia70, and TNF-α upregulated in experimental and human glaucoma71, 72. Moreover, the level of the TNF-α increased in the aqueous humor of glaucoma, and TNF-α gene polymorphisms have been correlated with POAG73, 74. Intravitreal TNF-α injection has a similar effect to the ocular hypertension glaucoma mouse model; blocking the TNF-α signaling could guard against the loss of RGCs71. TNF-α mediated RGC death in a caspase-independent event, but may mediate the insertion of Ca2+ -permeable AMPAR during excitotoxic injury and increase RGCs susceptibility to damage70, 75. Anti-inflammatory drugs’ targeting of the TNF-α signaling pathway has been fully explored, and the most promising one is Copolymer-1(Cop-1), which has been approved to treat multiple sclerosis (MS)76. Cop-1 binds to the relevant major histocompatibility complex proteins and leads to the activation of T suppressor cells, triggering a neuroprotective autoimmune response77. It has been demonstrated to protect the RGCs in vivo in the rat model of optic nerve crush77, in the high-IOP model78, and against glutamate-induced excitotoxicity79. 6. Anti-apoptotic strategies and gene therapy The apoptosis of the RGCs have been demonstrated in different kinds of animal models inducing RGCs loss. The apoptotic process can be triggered by various stimuli and involves intrinsic and extrinsic pathways, which are the subject of intense research in the quest for molecular targets to prevent RGC death in glaucoma. Apoptosis signal regulating kinase1 (ASK1) which is primarily expressed by RGC, has an important role in stress-induced RGC apoptosis80. Increased RGC survival and decreased degenerating axons in the optic nerve were observed in the ASK1 null mice81. The downstream factor of the ASK1 are called JNKs and have ten different isoforms, which display differential specificity toward their target82, playing a key role in the survival of the RGCs. Deleting one kind of JNK gene, the JNK3, did not prevent RGC loss in the ocular hypertension models; nonetheless, both eliminating the JNK2 and JNK3 showed a significant RGC-protective effect after injury83, 84. This may suggest that different JNK isoforms serve to compensate for deficiencies in this pathway to ensure RGC apoptosis. Generally, the anti-apoptotic Bcl-2 and pro-apoptotic Bax genes play an important role in the survival of the RGCs. Overexpression of either the Bcl-2 or Bcl-XL gene prevents RGC loss in optic nerve crush models85, 86. On the other hand, deleting the bax genes result in RGC neuroprotection when subjected to optic nerve crush models or in a genetic model of glaucoma87, 88. The common pathway that the intrinsic and the extrinsic apoptotic have is activating the caspase family, including caspase-9/3, caspase-8, so the inhibition of caspases to protect RGC is a promising strategy. Whether one uses an intraocular injection of caspase inhibitors or gene therapy to lower the expression of the caspases, they all show modest RGC protection32, 89. A combination of overexpression for both BIRC4/XIAP (a protein directly inhibiting caspase-3,7,9) and GDNF had a synergistic effect on the survival of axotomized RGCs90. Therefore, the strategy to target multiple anti-apoptotic pathways appears to be a promising method to prevent RGC loss in glaucoma. 7. Protein Misfolding More and more evidence has demonstrated that glaucoma is a progressive neurodegenerative disease32, 91, similar to Alzheimer’s in that both have aggregates of amyloid-β92. Glaucomatous retinopathy and visual field loss are more likely for people with AD91. Research has found that Aβ localizes with apoptotic RGCs in experimental glaucoma and induced RGC apoptosis93. Thus, obstructing the Aβ pathway provides a therapeutic avenue in glaucoma management. N-benzyloxycarbonyl-Val-Leu-leucinal (Z-VLL-CHO), a kind of β-secretase inhibitor, has been found to reduce RGC apoptosis both in vitro and in vivo94. Using the agents that target Aβ, including β-secretase inhibitors, Congo red, and anti-Aβ antibodies, is more effective than monotherapy93.Hot shock proteins (HSPs) is a group of molecular chaperones that helps to form and maintain the proper conformation of other proteins and prevent abnormal protein aggregating in the cell body95. Increased immunostaining of HSP 60 and HSP 27 has been confirmed in the glaucomatous eyes96; using geranylgeranylacetone (GGA) to promote the HSP 27 expression in the RGCs has reduced RGC loss in high-IOP animal models97. However, it is still poorly known how these HSPs function in the glaucoma. 8. Ischemia and drugs Research has shown that dysfunction of the microcirculation at the optic nerve head plays an important role in glaucoma, which may be caused inherently or by high IOP98. High levels of the endothein-1(ET-1) have been found in the aqueous humor of glaucoma patients99, and intravitreal injection of ET-1 could impair axonal transport in RGCs, which may constrict the microvasculature of the optic nerve head and retina100, 101. Calcium-channel blockers (CCBs) have antivasospastic and anti-ischemic effects. Lomerizine alleviates secondary degeneration of RGCs in an optic nerve crush model102, 103, while a study of the effect of nilvadipine, a calcium antagonist, showed that it slightly slowed the visual field progression, maintained the optic disc rim, and increased posterior choroidal circulation after three years of following up104. However, more attention should be paid to the potential of the CCBs’ reducing the optic nerve head perfusion pressure105. 9. Others Brimonidine, a drug previously shown to be neuroprotective in the laboratory, may have had a beneficial effect on visual function independent of IOP lowering in a low-pressure glaucoma treatment study106. L-N(6)-(1-iminoethyl)lysine 5-tetrazole amide, a prodrug of an inhibitor of inducible nitric oxide synthase, decreased generation of the nitric oxide at the optic nerve head could mitigate the RGC loss in a rat model of glaucoma107. Promising research areas in the future 1. Axonal transport failure and synaptic dysfunction For RGCs to survive, they and their target neuron cells in the lateral geniculate nucleus (LGN) must be successfully linked to exchange information and acquire sufficient neurotrophic factors. However, deficits in both anterograde and retrograde axonal transport in the optic nerve have been observed in animal glaucoma models and in human high-pressure secondary glaucoma108, 109. When subject to high IOP, retrograde transport of BDNF was impaired and dynein (a motor protein required for axonal transport) accumulated at the optic nerve head and retinal110, 111. More and more evidence suggests that the primary injury in the glaucoma is the deficit in retrograde axonal transport, which occurs early and progresses in a distal-to-proximal pattern112. More interestingly, the RGC axons and their presynaptic neurons persist in the colliculus well after transport fails. This means that restoration of transport along RGC axons might be an early therapeutic target for glaucoma112. Furthermore, detecting the damage of the axonal transport in the glaucoma will provide us with the clues for early diagnosis and the time for early intervention. The synaptic connectivity dysfunction may also be an early characteristic in glaucoma. Many studies have demonstrated that RGC dendritic thinning, reduced arbor complexity, and arbor retraction occurred prior to RGC shrinkage and axon atrophy113, 114. Reduced levels of c-fos, a marker of neuronal connectivity, happened early in ocular hypertension rats115, and morphological changes to the RGC dendrites related to a reduction in the spatial and temporal response to visual stimuli may underlie functional deficits in glaucoma116. Finding a way to detect the early damage of dendrites will provide us with an early clue to the diagnosis, and discovering methods to preserve the normal function of synaptic connectivity will offer promising strategies to treating glaucoma. 2. Glial cell system An interesting issue has occurred in the field of glaucoma neuroprotection: The traditional RGC-centric view of neurodegeneration in glaucoma has changed, acknowledging that other retinal- and optic-nerve cells actively contribute to RGC death. The neuron-glia interaction in the retinal and optic nerve head has received considerable attention. Astrocytes, Müller cells, and microglial cells in the retinal nerve were proposed to play both protective and deleterious roles in glaucoma. They are also the key resident immune cells in the retina and optic nerve, thus playing a key role in immune response in glaucoma. Astrocytes play a key role in the remodeling of the optic nerve head during glaucomatous damage117, which is confirmed by the fact that astrocytes proliferated at the optic nerve head and astrocytosis has been observed in the LGN and visual cortex in human glaucoma and animal glaucoma models118, 119. Recent studies found that the astrocytes at the myelination transition zone, which express phagocytic marker Mac-2, enhanced the capability of phagocytose RGC axonal processes, suggesting that this degradative pathway for axons might contribute to glaucomatous neurodegeneration120. In addition to modulating the optic nerve head environment, astrocytes are also known to produce neurotoxic molecules when subject to injury, which in turn leads to RGC death121, 122. Müller cells, the most abundant retinal glia cell type, are among the first responders following IOP increase. Reactive Müller cells in glaucoma could increase the susceptibility of RGCs to stress signals and contribute to disease progression123. Deficit in the potassium buffering system, water clearance, or production of antioxidant molecules by Müller cells could impair RGC function in glaucoma70. Moreover, Müller cells also take part in the oxidative stress in the RGC70. One exciting discovery is that Müller cells are endowed with stem cell properties and are being explored as a potential source of neural replacement and transplantation in the injured visual system124, 125. Nonetheless, the exact role the Müller cells play in the glaucoma and what people could do to modulate the Müller cell to have a protective role are still vastly underexplored. Microglia are specialized innate immune cells that reside in the retina and the brain. Microglia could change from a quiescent state to an activated state when the microenvironment changes, with their morphology and cell-surface molecules transforming, leading to the clearance of toxic debris and release of neurotrophic and anti-inflammatory factors126-128. However, over-activation of the microglia results in detrimental effects on neurons129, 130. Reactive microglia has been observed in the retina and optic nerves from axotomized eyes during ocular hypertension and human glaucoma131. Inhibition of microglia over-activation with minocycline delayed RGC death in the animal models132. However, more research is needed to explore in-depth the potential therapeutic role microglia play in glaucoma. 3. Stem cells Neuroprotection aims to halt or slow the death of RGCs and their axons in the glaucoma, but how does one compensate for the loss of RGCs? Stem cell transplantation may provide a promising new avenue for treating glaucoma. There are two prospects for cell transplantation-based treatment modalities: neuroprotection and RGC replacement. Interestingly, research shows that the transplantation of stem cells secretes high levels of neurotrophins, including NTFs, which have a protective effect on RGCs in experimental glaucoma133, 134. There has been great progress in the regeneration of RGCs, which provides encouragement that RGC replacement may be possible135-137. However, obstacles remain, including how to acquire functional RGCs from the stem cells, how to optimize for maximal engraftment and integration into the host retina, and how to promote the excellent synaptic connection with the brain. All of these questions require a great deal more research. Challenges and assessment of the neuroprotection strategies There is great diversity to the molecular signals in the glaucoma, and it is highly plausible that different molecular pathways are activated at different stages of glaucoma onset and progression32. Identifying the exact issues that occur at the particular stages is essential if one is to combine the various therapies targeting different mechanisms. Every RGC should also have its own receptor profiles and unique individual characteristics, allowing each one to respond differently to injury. This means that apoptosis can be triggered in different ganglion cells for different reasons and at different times; as a result, an agent with only a single mode of action may not completely protect all the RGCs. Understanding these issues better will help us develop personalized therapies for neuroprotection in glaucoma. What is more, it is still difficult to detect the progressive glaucomatous injury, although techniques for imaging the living RGCs have developed in animals138, 139. At present, the endpoint of the study depends on measuring the functional injury, such as the visual field, but it does not take into account the structural alterations in the optic nerve or retinal nerve fiber layer. Moreover, the duration of clinical trials of neuroprotection may be longer as glaucomatous optic neuropathy is a slowly progressive disease. R.N. Weinreb suggested a criterion for evaluating a glaucoma neuroprotective agent’s potential8: “First, the drug should have a specific receptor target in the retinal or optic nerve. Second, the drug must reach the retinal or optic nerve in pharmacologically effective concentrations. Third, evidence must be obtained in animal models that activation of the target triggers pathways that enhance neuronal survival or decrease neuronal damage. Fourth, the neuroprotective activity must be demonstrated in randomized, controlled, clinical trials in humans”. Based on the above criterion, we should consider the following issues before we find an effective neuroprotective agent, the exact pathogenesis of the glaucoma, the optimal glaucoma animal models, and an appropriate clinical assessment system. CONCLUSIONS Although there is strong or convincing laboratory evidence that several drugs provide neuroprotection in glaucoma, only a minority of these investigations has led to approved therapies, and the road to the implementation of neuroprotection in glaucoma is still long. The evidence from randomized clinical trials is still lacking. A considerable investment in genetic studies and molecular investigations may yield more progress. REFERENCE 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol 2006;90:262-7. Quigley HA. Glaucoma. Lancet 2011;377:1367-77. Kass MA, Heuer DK, Higginbotham EJ, Johnson CA, Keltner JL, Miller JP, et al. The Ocular Hypertension Treatment Study: a randomized trial determines thattopical ocular hypotensive medication delays or prevents the onset of primaryopen-angle glaucoma. Arch Ophthalmol 2002;120:701-13; discussion 829-30. Lichter PR, Musch DC, Gillespie BW, Guire KE, Janz NK, Wren PA, et al. Interim clinical outcomes in the Collaborative Initial Glaucoma Treatment Studycomparing initial treatment randomized to medications or surgery. Ophthalmology 2001;108:1943-53. Musch DC, Gillespie BW, Lichter PR, Niziol LM, Janz NK. Visual field progression in the Collaborative Initial Glaucoma Treatment Studythe impact of treatment and other baseline factors. Ophthalmology 2009;116:200-7. Leske MC, Heijl A, Hussein M, Bengtsson B, Hyman L, Komaroff E. Factors for glaucoma progression and the effect of treatment: the early manifest glaucoma trial. Arch Ophthalmol 2003;121:48-56. Chang EE, Goldberg JL. Glaucoma 2.0: neuroprotection, neuroregeneration, neuroenhancement. Ophthalmology 2012;119:979-86. Weinreb RN. Glaucoma neuroprotection: What is it? Why is it needed. Can J Ophthalmol 2007;42:396-8. Osborne NN. Recent clinical findings with memantine should not mean that the idea ofneuroprotection in glaucoma is abandoned. Acta Ophthalmol 2009;87:450-4. Teng KK, Hempstead BL. Neurotrophins and their receptors: signaling trios in complex biological systems. Cell Mol Life Sci 2004;61:35-48. Miller FD, Kaplan DR. Neurotrophin signalling pathways regulating neuronal 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. apoptosis. Cell Mol Life Sci 2001;58:1045-53. Lebrun-Julien F, Bertrand MJ, De Backer O, Stellwagen D, Morales CR, Di PA, et al. ProNGF induces TNFalpha-dependent death of retinal ganglion cells through ap75NTR non-cell-autonomous signaling pathway. Proc Natl Acad Sci U S A 2010;107:3817-22. Pasutto F, Matsumoto T, Mardin CY, Sticht H, Brandstatter JH, Michels-Rautenstrauss K, et al. Heterozygous NTF4 mutations impairing neurotrophin-4 signaling in patients withprimary open-angle glaucoma. Am J Hum Genet 2009;85:447-56. Lambiase A, Aloe L, Centofanti M, Parisi V, Mantelli F, Colafrancesco V, et al. Experimental and clinical evidence of neuroprotection by nerve growth factor eye drops: Implications for glaucoma. Proc Natl Acad Sci U S A 2009. Martin KR, Quigley HA, Zack DJ, Levkovitch-Verbin H, Kielczewski J, Valenta D, et al. Gene therapy with brain-derived neurotrophic factor as a protection: retinalganglion cells in a rat glaucoma model. Invest Ophthalmol Vis Sci 2003;44:4357-65. Chen H, Weber AJ. BDNF enhances retinal ganglion cell survival in cats with optic nerve damage. Invest Ophthalmol Vis Sci 2001;42:966-74. Koeberle PD, Ball AK. Neurturin enhances the survival of axotomized retinal ganglion cells in vivo:combined effects with glial cell line-derived neurotrophic factor andbrain-derived neurotrophic factor. Neuroscience 2002;110:555-67. Arancio O, Chao MV. Neurotrophins, synaptic plasticity and dementia. Curr Opin Neurobiol 2007;17:325-30. Schmidt H, Rathjen FG. Signalling mechanisms regulating axonal branching in vivo. Bioessays 2010;32:977-85. Weber AJ, Harman CD. BDNF preserves the dendritic morphology of alpha and beta ganglion cells in thecat retina after optic nerve injury. Invest Ophthalmol Vis Sci 2008;49:2456-63. Chun MH, Ju WK, Kim KY, Lee MY, Hofmann HD, Kirsch M, et al. Upregulation of ciliary neurotrophic factor in reactive Muller cells in the ratretina following optic nerve transection. Brain Res 2000;868:358-62. Wu Q, Zhang M, Song BW, Lu B, Hu P. Expression of ciliary neurotrophic factor after induction of ocular hypertension in the retina of rats. Chin Med J (Engl) 2007;120:1825-9. Pease ME, Zack DJ, Berlinicke C, Bloom K, Cone F, Wang Y, et al. Effect of CNTF on retinal ganglion cell survival in experimental glaucoma. Invest Ophthalmol Vis Sci 2009;50:2194-200. Ji JZ, Elyaman W, Yip HK, Lee VW, Yick LW, Hugon J, et al. CNTF promotes survival of retinal ganglion cells after induction of ocularhypertension in rats: the possible involvement of STAT3 pathway. Eur J Neurosci 2004;19:265-72. Leaver SG, Cui Q, Plant GW, Arulpragasam A, Hisheh S, Verhaagen J, et al. AAV-mediated expression of CNTF promotes long-term survival and regeneration ofadult rat retinal ganglion cells. Gene Ther 2006;13:1328-41. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. McGill TJ, Prusky GT, Douglas RM, Yasumura D, Matthes MT, Nune G, et al. Intraocular CNTF reduces vision in normal rats in a dose-dependent manner. Invest Ophthalmol Vis Sci 2007;48:5756-66. Skaper SD. Peptide mimetics of neurotrophins and their receptors. Curr Pharm Des 2011;17:2704-18. Gozes I. Neuroprotective peptide drug delivery and development: potential newtherapeutics. Trends Neurosci 2001;24:700-5. Lagreze WA, Pielen A, Steingart R, Schlunck G, Hofmann HD, Gozes I, et al. The peptides ADNF-9 and NAP increase survival and neurite outgrowth of ratretinal ganglion cells in vitro. Invest Ophthalmol Vis Sci 2005;46:933-8. Cheng L, Sapieha P, Kittlerova P, Hauswirth WW, Di PA. TrkB gene transfer protects retinal ganglion cells from axotomy-induced death in vivo. J Neurosci 2002;22:3977-86. Logan A, Ahmed Z, Baird A, Gonzalez AM, Berry M. Neurotrophic factor synergy is required for neuronal survival and disinhibitedaxon regeneration after CNS injury. Brain 2006;129:490-502. Almasieh M, Wilson AM, Morquette B, Cueva VJL, Di PA. The molecular basis of retinal ganglion cell death in glaucoma. Prog Retin Eye Res 2012;31:152-81. Seki M, Lipton SA. Targeting excitotoxic/free radical signaling pathways for therapeuticintervention in glaucoma. Prog Brain Res 2008;173:495-510. Schuettauf F, Stein T, Choragiewicz TJ, Rejdak R, Bolz S, Zurakowski D, et al. Caspase inhibitors protect against NMDA-mediated retinal ganglion cell death. Clin Experiment Ophthalmol 2011;39:545-54. Dong CJ, Guo Y, Agey P, Wheeler L, Hare WA. Alpha2 adrenergic modulation of NMDA receptor function as a major mechanism ofRGC protection in experimental glaucoma and retinal excitotoxicity. Invest Ophthalmol Vis Sci 2008;49:4515-22. Wamsley S, Gabelt BT, Dahl DB, Case GL, Sherwood RW, May CA, et al. Vitreous glutamate concentration and axon loss in monkeys with experimentalglaucoma. Arch Ophthalmol 2005;123:64-70. Honkanen RA, Baruah S, Zimmerman MB, Khanna CL, Weaver YK, Narkiewicz J, et al. Vitreous amino acid concentrations in patients with glaucoma undergoingvitrectomy. Arch Ophthalmol 2003;121:183-8. Hartwick AT, Hamilton CM, Baldridge WH. Glutamatergic calcium dynamics and deregulation of rat retinal ganglion cells. J Physiol 2008;586:3425-46. Furukawa Y, Okada M, Akaike N, Hayashi T, Nabekura J. Reduction of voltage-dependent magnesium block of N-methyl-D-aspartatereceptor-mediated current by in vivo axonal injury. Neuroscience 2000;96:385-92. Hare WA, WoldeMussie E, Lai RK, Ton H, Ruiz G, Chun T, et al. Efficacy and safety of memantine treatment for reduction of changes associatedwith experimental glaucoma in monkey, I: Functional measures. Invest Ophthalmol Vis Sci 2004;45:2625-39. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. Yucel YH, Gupta N, Zhang Q, Mizisin AP, Kalichman MW, Weinreb RN. Memantine protects neurons from shrinkage in the lateral geniculate nucleus inexperimental glaucoma. Arch Ophthalmol 2006;124:217-25. Osborne NN. Recent clinical findings with memantine should not mean that the idea ofneuroprotection in glaucoma is abandoned. Acta Ophthalmol 2009;87:450-4. Russo R, Cavaliere F, Berliocchi L, Nucci C, Gliozzi M, Mazzei C, et al. Modulation of pro-survival and death-associated pathways under retinalischemia/reperfusion: effects of NMDA receptor blockade. J Neurochem 2008;107:1347-57. Guo L, Salt TE, Maass A, Luong V, Moss SE, Fitzke FW, et al. Assessment of neuroprotective effects of glutamate modulation on glaucoma-relatedretinal ganglion cell apoptosis in vivo. Invest Ophthalmol Vis Sci 2006;47:626-33. Cross JV, Templeton DJ. Thiol oxidation of cell signaling proteins: Controlling an apoptotic equilibrium. J Cell Biochem 2004;93:104-11. Andersen JK. Oxidative stress in neurodegeneration: cause or consequence. Nat Med 2004;10 Suppl:S18-25. Kanamori A, Catrinescu MM, Kanamori N, Mears KA, Beaubien R, Levin LA. Superoxide is an associated signal for apoptosis in axonal injury. Brain 2010;133:2612-25. Moreno MC, Campanelli J, Sande P, Sanez DA, Keller SMI, Rosenstein RE. Retinal oxidative stress induced by high intraocular pressure. Free Radic Biol Med 2004;37:803-12. Yuki K, Ozawa Y, Yoshida T, Kurihara T, Hirasawa M, Ozeki N, et al. Retinal ganglion cell loss in superoxide dismutase 1 deficiency. Invest Ophthalmol Vis Sci 2011;52:4143-50. Swanson KI, Schlieve CR, Lieven CJ, Levin LA. Neuroprotective effect of sulfhydryl reduction in a rat optic nerve crush model. Invest Ophthalmol Vis Sci 2005;46:3737-41. Tezel G, Yang X, Cai J. Proteomic identification of oxidatively modified retinal proteins in a chronicpressure-induced rat model of glaucoma. Invest Ophthalmol Vis Sci 2005;46:3177-87. Caprioli J, Munemasa Y, Kwong JM, Piri N. Overexpression of thioredoxins 1 and 2 increases retinal ganglion cell survivalafter pharmacologically induced oxidative stress, optic nerve transection, and inexperimental glaucoma. Trans Am Ophthalmol Soc 2009;107:161-5. Siu AW, Ortiz GG, Benitez-King G, To CH, Reiter RJ. Effects of melatonin on the nitric oxide treated retina. Br J Ophthalmol 2004;88:1078-81. Yancheva S, Ihl R, Nikolova G, Panayotov P, Schlaefke S, Hoerr R. Ginkgo biloba extract EGb 761(R), donepezil or both combined in the treatment of Alzheimer's disease with neuropsychiatric features: a randomised, double-blind,exploratory trial. Aging Ment Health 2009;13:183-90. Quaranta L, Bettelli S, Uva MG, Semeraro F, Turano R, Gandolfo E. Effect of Ginkgo biloba extract on preexisting visual field damage in normaltension 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. glaucoma. Ophthalmology 2003;110:359-62; discussion 362-4. Hirooka K, Tokuda M, Miyamoto O, Itano T, Baba T, Shiraga F. The Ginkgo biloba extract (EGb 761) provides a neuroprotective effect on retinal ganglion cells in a rat model of chronic glaucoma. Curr Eye Res 2004;28:153-7. Thiagarajan G, Chandani S, Harinarayana RS, Samuni AM, Chandrasekaran K, Balasubramanian D. Molecular and cellular assessment of ginkgo biloba extract as a possibleophthalmic drug. Exp Eye Res 2002;75:421-30. Cheung ZH, Yip HK, Wu W, So KF. Axotomy induces cytochrome c release in retinal ganglion cells. Neuroreport 2003;14:279-82. Wang L, Dong J, Cull G, Fortune B, Cioffi GA. Varicosities of intraretinal ganglion cell axons in human and nonhuman primates. Invest Ophthalmol Vis Sci 2003;44:2-9. 3rd AA. CNS energy metabolism as related to function. Brain Res Brain Res Rev 2000;34:42-68. Baltan S, Inman DM, Danilov CA, Morrison RS, Calkins DJ, Horner PJ. Metabolic vulnerability disposes retinal ganglion cell axons to dysfunction in a model of glaucomatous degeneration. J Neurosci 2010;30:5644-52. Hu H, Lu W, Zhang M, Zhang X, Argall AJ, Patel S, et al. Stimulation of the P2X7 receptor kills rat retinal ganglion cells in vivo. Exp Eye Res 2010;91:425-32. Schober MS, Chidlow G, Wood JP, Casson RJ. Bioenergetic-based neuroprotection and glaucoma. Clin Experiment Ophthalmol 2008;36:377-85. Lee S, Van Bergen NJ, Kong GY, Chrysostomou V, Waugh HS, O'Neill EC, et al. Mitochondrial dysfunction in glaucoma and emerging bioenergetic therapies. Exp Eye Res 2011;93:204-12. Jones AW, Yao Z, Vicencio JM, Karkucinska-Wieckowska A, Szabadkai G. PGC-1 family coactivators and cell fate: roles in cancer, neurodegeneration,cardiovascular disease and retrograde mitochondria-nucleus signalling. Mitochondrion 2012;12:86-99. Nakajima Y, Inokuchi Y, Nishi M, Shimazawa M, Otsubo K, Hara H. Coenzyme Q10 protects retinal cells against oxidative stress in vitro and invivo. Brain Res 2008;1226:226-33. Nucci C, Tartaglione R, Cerulli A, Mancino R, Spano A, Cavaliere F, et al. Retinal damage caused by high intraocular pressure-induced transient ischemia is prevented by coenzyme Q10 in rat. Int Rev Neurobiol 2007;82:397-406. Tezel G. Immune regulation toward immunomodulation for neuroprotection in glaucoma.LID S1471-4892(12)00195-6 [pii]LID 10.1016/j.coph.2012.09.013 [doi]. Curr Opin Pharmacol 2012. Tezel G. The role of glia, mitochondria, and the immune system in glaucoma. Invest Ophthalmol Vis Sci 2009;50:1001-12. Lebrun-Julien F, Duplan L, Pernet V, Osswald I, Sapieha P, Bourgeois P, et al. Excitotoxic death of retinal neurons in vivo occurs via a non-cell-autonomousmechanism. J Neurosci 2009;29:5536-45. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. Nakazawa T, Nakazawa C, Matsubara A, Noda K, Hisatomi T, She H, et al. Tumor necrosis factor-alpha mediates oligodendrocyte death and delayed retinalganglion cell loss in a mouse model of glaucoma. J Neurosci 2006;26:12633-41. Tezel G, Li LY, Patil RV, Wax MB. TNF-alpha and TNF-alpha receptor-1 in the retina of normal and glaucomatous eyes. Invest Ophthalmol Vis Sci 2001;42:1787-94. Balaiya S, Edwards J, Tillis T, Khetpal V, Chalam KV. Tumor necrosis factor-alpha (TNF-alpha) levels in aqueous humor of primary openangle glaucoma. Clin Ophthalmol 2011;5:553-6. Bozkurt B, Mesci L, Irkec M, Ozdag BB, Sanal O, Arslan U, et al. Association of tumour necrosis factor-alpha -308 G/A polymorphism with primaryopen-angle glaucoma. Clin Experiment Ophthalmol 2012;40:e156-62. Tezel G, Yang X. Caspase-independent component of retinal ganglion cell death, in vitro. Invest Ophthalmol Vis Sci 2004;45:4049-59. Zang Y, Hong J, Robinson R, Li S, Rivera VM, Zhang JZ. Immune regulatory properties and interactions of copolymer-I and beta-interferon 1a in multiple sclerosis. J Neuroimmunol 2003;137:144-53. Kipnis J, Yoles E, Porat Z, Cohen A, Mor F, Sela M, et al. T cell immunity to copolymer 1 confers neuroprotection on the damaged opticnerve: possible therapy for optic neuropathies. Proc Natl Acad Sci U S A 2000;97:7446-51. Bakalash S, Kessler A, Mizrahi T, Nussenblatt R, Schwartz M. Antigenic specificity of immunoprotective therapeutic vaccination for glaucoma. Invest Ophthalmol Vis Sci 2003;44:3374-81. Schori H, Kipnis J, Yoles E, WoldeMussie E, Ruiz G, Wheeler LA, et al. Vaccination for protection of retinal ganglion cells against death from glutamatecytotoxicity and ocular hypertension: implications for glaucoma. Proc Natl Acad Sci U S A 2001;98:3398-403. Hattori K, Naguro I, Runchel C, Ichijo H. The roles of ASK family proteins in stress responses and diseases. Cell Commun Signal 2009;7:9. Harada C, Namekata K, Guo X, Yoshida H, Mitamura Y, Matsumoto Y, et al. ASK1 deficiency attenuates neural cell death in GLAST-deficient mice, a model of normal tension glaucoma. Cell Death Differ 2010;17:1751-9. Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature 2001;410:37-40. Quigley HA, Cone FE, Gelman SE, Yang Z, Son JL, Oglesby EN, et al. Lack of neuroprotection against experimental glaucoma in c-Jun N-terminal kinase 3 knockout mice. Exp Eye Res 2011;92:299-305. Fernandes KA, Harder JM, Fornarola LB, Freeman RS, Clark AF, Pang IH, et al. JNK2 and JNK3 are major regulators of axonal injury-induced retinal ganglion celldeath. Neurobiol Dis 2012;46:393-401. Malik JM, Shevtsova Z, Bahr M, Kugler S. Long-term in vivo inhibition of CNS neurodegeneration by Bcl-XL gene transfer. Mol Ther 2005;11:373-81. Planchamp V, Bermel C, Tonges L, Ostendorf T, Kugler S, Reed JC, et al. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. BAG1 promotes axonal outgrowth and regeneration in vivo via Raf-1 and reductionof ROCK activity. Brain 2008;131:2606-19. Libby RT, Li Y, Savinova OV, Barter J, Smith RS, Nickells RW, et al. Susceptibility to neurodegeneration in a glaucoma is modified by Bax gene dosage. PLoS Genet 2005;1:17-26. Li Y, Schlamp CL, Poulsen KP, Nickells RW. Bax-dependent and independent pathways of retinal ganglion cell death induced by different damaging stimuli. Exp Eye Res 2000;71:209-13. Lingor P, Koeberle P, Kugler S, Bahr M. Down-regulation of apoptosis mediators by RNAi inhibits axotomy-induced retinalganglion cell death in vivo. Brain 2005;128:550-8. Straten G, Schmeer C, Kretz A, Gerhardt E, Kugler S, Schulz JB, et al. Potential synergistic protection of retinal ganglion cells from axotomy-inducedapoptosis by adenoviral administration of glial cell line-derived neurotrophicfactor and X-chromosome-linked inhibitor of apoptosis. Neurobiol Dis 2002;11:123-33. Wostyn P, Audenaert K, De Deyn PP. Alzheimer's disease and glaucoma: is there a causal relationship. Br J Ophthalmol 2009;93:1557-9. Goldblum D, Kipfer-Kauer A, Sarra GM, Wolf S, Frueh BE. Distribution of amyloid precursor protein and amyloid-beta immunoreactivity inDBA/2J glaucomatous mouse retinas. Invest Ophthalmol Vis Sci 2007;48:5085-90. Guo L, Salt TE, Luong V, Wood N, Cheung W, Maass A, et al. Targeting amyloid-beta in glaucoma treatment. Proc Natl Acad Sci U S A 2007;104:13444-9. Yamamoto R, Yoneda S, Hara H. Neuroprotective effects of beta-secretase inhibitors against rat retinal ganglioncell death. Neurosci Lett 2004;370:61-4. Soti C, Nagy E, Giricz Z, Vigh L, Csermely P, Ferdinandy P. Heat shock proteins as emerging therapeutic targets. Br J Pharmacol 2005;146:769-80. Tezel G, Hernandez R, Wax MB. Immunostaining of heat shock proteins in the retina and optic nerve head ofnormal and glaucomatous eyes. Arch Ophthalmol 2000;118:511-8. Ishii Y, Kwong JM, Caprioli J. Retinal ganglion cell protection with geranylgeranylacetone, a heat shock proteininducer, in a rat glaucoma model. Invest Ophthalmol Vis Sci 2003;44:1982-92. Hafez AS, Bizzarro RL, Lesk MR. Evaluation of optic nerve head and peripapillary retinal blood flow in glaucomapatients, ocular hypertensives, and normal subjects. Am J Ophthalmol 2003;136:1022-31. Choritz L, Machert M, Thieme H. Correlation of endothelin-1 concentration in aqueous humor with intraocularpressure in primary open angle and pseudoexfoliation glaucoma. Invest Ophthalmol Vis Sci 2012;53:7336-42. Stokely ME, Brady ST, Yorio T. Effects of endothelin-1 on components of anterograde axonal transport in opticnerve. Invest Ophthalmol Vis Sci 2002;43:3223-30. Sasaoka M, Taniguchi T, Shimazawa M, Ishida N, Shimazaki A, Hara H. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. Intravitreal injection of endothelin-1 caused optic nerve damage following toocular hypoperfusion in rabbits. Exp Eye Res 2006;83:629-37. Fitzgerald M, Payne SC, Bartlett CA, Evill L, Harvey AR, Dunlop SA. Secondary retinal ganglion cell death and the neuroprotective effects of thecalcium channel blocker lomerizine. Invest Ophthalmol Vis Sci 2009;50:5456-62. Karim Z, Sawada A, Kawakami H, Yamamoto T, Taniguchi T. A new calcium channel antagonist, lomerizine, alleviates secondary retinalganglion cell death after optic nerve injury in the rat. Curr Eye Res 2006;31:273-83. Koseki N, Araie M, Tomidokoro A, Nagahara M, Hasegawa T, Tamaki Y, et al. A placebo-controlled 3-year study of a calcium blocker on visual field and ocularcirculation in glaucoma with low-normal pressure. Ophthalmology 2008;115:2049-57. Costa VP, Arcieri ES, Harris A. Blood pressure and glaucoma. Br J Ophthalmol 2009;93:1276-82. Krupin T, Liebmann JM, Greenfield DS, Ritch R, Gardiner S. A randomized trial of brimonidine versus timolol in preserving visual function:results from the Low-Pressure Glaucoma Treatment Study. Am J Ophthalmol 2011;151:671-81. Neufeld AH, Das S, Vora S, Gachie E, Kawai S, Manning PT, et al. A prodrug of a selective inhibitor of inducible nitric oxide synthase isneuroprotective in the rat model of glaucoma. J Glaucoma 2002;11:221-5. Salinas-Navarro M, Alarcon-Martinez L, Valiente-Soriano FJ, Jimenez-Lopez M, Mayor-Torroglosa S, Aviles-Trigueros M, et al. Ocular hypertension impairs optic nerve axonal transport leading to progressiveretinal ganglion cell degeneration. Exp Eye Res 2010;90:168-83. Knox DL, Eagle RC Jr, Green WR. Optic nerve hydropic axonal degeneration and blocked retrograde axoplasmictransport: histopathologic features in human high-pressure secondary glaucoma. Arch Ophthalmol 2007;125:347-53. Pease ME, McKinnon SJ, Quigley HA, Kerrigan-Baumrind LA, Zack DJ. Obstructed axonal transport of BDNF and its receptor TrkB in experimentalglaucoma. Invest Ophthalmol Vis Sci 2000;41:764-74. Martin KR, Quigley HA, Valenta D, Kielczewski J, Pease ME. Optic nerve dynein motor protein distribution changes with intraocular pressureelevation in a rat model of glaucoma. Exp Eye Res 2006;83:255-62. Crish SD, Sappington RM, Inman DM, Horner PJ, Calkins DJ. Distal axonopathy with structural persistence in glaucomatous neurodegeneration. Proc Natl Acad Sci U S A 2010;107:5196-201. Morgan JE, Datta AV, Erichsen JT, Albon J, Boulton ME. Retinal ganglion cell remodelling in experimental glaucoma. Adv Exp Med Biol 2006;572:397-402. Liu M, Duggan J, Salt TE, Cordeiro MF. Dendritic changes in visual pathways in glaucoma and other neurodegenerativeconditions. Exp Eye Res 2011;92:244-50. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. Fu QL, Li X, Shi J, Xu G, Wen W, Lee DH, et al. Synaptic degeneration of retinal ganglion cells in a rat ocular hypertensionglaucoma model. Cell Mol Neurobiol 2009;29:575-81. Weber AJ, Harman CD. Structure-function relations of parasol cells in the normal and glaucomatousprimate retina. Invest Ophthalmol Vis Sci 2005;46:3197-207. Hernandez MR. The optic nerve head in glaucoma: role of astrocytes in tissue remodeling. Prog Retin Eye Res 2000;19:297-321. Johnson EC, Deppmeier LM, Wentzien SK, Hsu I, Morrison JC. Chronology of optic nerve head and retinal responses to elevated intraocularpressure. Invest Ophthalmol Vis Sci 2000;41:431-42. Lam D, Jim J, To E, Rasmussen C, Kaufman PL, Matsubara J. Astrocyte and microglial activation in the lateral geniculate nucleus and visual cortex of glaucomatous and optic nerve transected primates. Mol Vis 2009;15:2217-29. Nguyen JV, Soto I, Kim KY, Bushong EA, Oglesby E, Valiente-Soriano FJ, et al. Myelination transition zone astrocytes are constitutively phagocytic and havesynuclein dependent reactivity in glaucoma. Proc Natl Acad Sci U S A 2011;108:1176-81. Johnson EC, Doser TA, Cepurna WO, Dyck JA, Jia L, Guo Y, et al. Cell proliferation and interleukin-6-type cytokine signaling are implicated bygene expression responses in early optic nerve head injury in rat glaucoma. Invest Ophthalmol Vis Sci 2011;52:504-18. Yan X, Tezel G, Wax MB, Edward DP. Matrix metalloproteinases and tumor necrosis factor alpha in glaucomatous opticnerve head. Arch Ophthalmol 2000;118:666-73. Bringmann A, Iandiev I, Pannicke T, Wurm A, Hollborn M, Wiedemann P, et al. Cellular signaling and factors involved in Muller cell gliosis: neuroprotectiveand detrimental effects. Prog Retin Eye Res 2009;28:423-51. Bernardos RL, Barthel LK, Meyers JR, Raymond PA. Late-stage neuronal progenitors in the retina are radial Muller glia thatfunction as retinal stem cells. J Neurosci 2007;27:7028-40. Bull ND, Limb GA, Martin KR. Human Muller stem cell (MIO-M1) transplantation in a rat model of glaucoma:survival, differentiation, and integration. Invest Ophthalmol Vis Sci 2008;49:3449-56. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma invivo. Science 2005;308:1314-8. Cho BP, Song DY, Sugama S, Shin DH, Shimizu Y, Kim SS, et al. Pathological dynamics of activated microglia following medial forebrain bundletransection. Glia 2006;53:92-102. Schwartz M, Butovsky O, Bruck W, Hanisch UK. Microglial phenotype: is the commitment reversible. Trends Neurosci 2006;29:68-74. Sappington RM, Calkins DJ. Contribution of TRPV1 to microglia-derived IL-6 and NFkappaB translocation withelevated hydrostatic pressure. Invest Ophthalmol Vis Sci 2008;49:3004-17. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. Lam TT, Kwong JM, Tso MO. Early glial responses after acute elevated intraocular pressure in rats. Invest Ophthalmol Vis Sci 2003;44:638-45. Yuan L, Neufeld AH. Activated microglia in the human glaucomatous optic nerve head. J Neurosci Res 2001;64:523-32. Bosco A, Inman DM, Steele MR, Wu G, Soto I, Marsh-Armstrong N, et al. Reduced retina microglial activation and improved optic nerve integrity withminocycline treatment in the DBA/2J mouse model of glaucoma. Invest Ophthalmol Vis Sci 2008;49:1437-46. Bull ND, Irvine KA, Franklin RJ, Martin KR. Transplanted oligodendrocyte precursor cells reduce neurodegeneration in a model of glaucoma. Invest Ophthalmol Vis Sci 2009;50:4244-53. Crigler L, Robey RC, Asawachaicharn A, Gaupp D, Phinney DG. Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp Neurol 2006;198:54-64. MacLaren RE, Pearson RA, MacNeil A, Douglas RH, Salt TE, Akimoto M, et al. Retinal repair by transplantation of photoreceptor precursors. Nature 2006;444:203-7. Lamba DA, McUsic A, Hirata RK, Wang PR, Russell D, Reh TA. Generation, purification and transplantation of photoreceptors derived from humaninduced pluripotent stem cells. PLOS ONE 2010;5:e8763. Lamba DA, Gust J, Reh TA. Transplantation of human embryonic stem cell-derived photoreceptors restores somevisual function in Crx-deficient mice. CELL STEM CELL 2009;4:73-9. Schmitz-Valckenberg S, Guo L, Maass A, Cheung W, Vugler A, Moss SE, et al. Real-time in vivo imaging of retinal cell apoptosis after laser exposure. Invest Ophthalmol Vis Sci 2008;49:2773-80. Barnett EM, Zhang X, Maxwell D, Chang Q, Piwnica-Worms D. Single-cell imaging of retinal ganglion cell apoptosis with a cell-penetrating,activatable peptide probe in an in vivo glaucoma model. Proc Natl Acad Sci U S A 2009;106:9391-6. Figure1 Multiple pathogenic mechanisms that result in axonal degeneration and RGC death in glaucoma