Next Article in Journal
Machine Learning Quantified Tumor-Stroma Ratio Is an Independent Prognosticator in Muscle-Invasive Bladder Cancer
Next Article in Special Issue
Immune Functions of Astrocytes in Viral Neuroinfections
Previous Article in Journal
Synthesis, In Silico Study, Antibacterial and Antifungal Activities of N-phenylbenzamides
Previous Article in Special Issue
Age-Related Changes in Neurons and Satellite Glial Cells in Mouse Dorsal Root Ganglia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Astrocyte Immune Functions and Glaucoma

by
Youichi Shinozaki
1,2,
Kenji Kashiwagi
3 and
Schuichi Koizumi
1,2,*
1
Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
2
Interdisciplinary Brain-Immune Research Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
3
Department of Ophthalmology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2747; https://doi.org/10.3390/ijms24032747
Submission received: 29 December 2022 / Revised: 26 January 2023 / Accepted: 26 January 2023 / Published: 1 February 2023
(This article belongs to the Special Issue Immune Functions of Astrocytes)

Abstract

:
Astrocytes, a non-neuronal glial cell type in the nervous system, are essential for regulating physiological functions of the central nervous system. In various injuries and diseases of the central nervous system, astrocytes often change their phenotypes into neurotoxic ones that participate in pro-inflammatory responses (hereafter referred to as “immune functions”). Such astrocytic immune functions are not only limited to brain diseases but are also found in ocular neurodegenerative diseases such as glaucoma, a retinal neurodegenerative disease that is the leading cause of blindness worldwide. The eye has two astrocyte-lineage cells: astrocytes and Müller cells. They maintain the physiological environment of the retina and optic nerve, thereby controlling visual function. Dysfunction of astrocyte-lineage cells may be involved in the onset and progression of glaucoma. These cells become reactive in glaucoma patients, and animal studies have suggested that their immune responses may be linked to glaucoma-related events: tissue remodeling, neuronal death, and infiltration of peripheral immune cells. In this review, we discuss the role of the immune functions of astrocyte-lineage cells in the pathogenesis of glaucoma.

1. Introduction

Astrocytes are a subtype of glial cells in the central nervous system. Glial cells are non-neuronal cell types with essential roles in regulating physiological brain functions and are divided into several types, including oligodendrocytes, microglia, and astrocytes [1]. They are not only limited to the brain but are also distributed in the peripheral nervous system, including the eye. The retina has two types of astrocyte-lineage cells: astrocytes and retina-specific Müller cells. Astrocytes are highly plastic cells and change their phenotypes into either neurodegenerative or neuroprotective ones under pathological conditions [2,3]. Astrocytes respond to inflammatory signals and can trigger inflammation by changing their phenotype into a neurodegenerative one. The neurodegenerative astrocytes are an important driver of various neurological diseases, including Alzheimer’s disease [4], Parkinson’s disease [5], Huntington’s disease [6], amyotrophic lateral sclerosis [7], and multiple sclerosis [8]. Various factors induce neurotoxic astrocytes, including lipopolysaccharide [9], normal brain aging [10], human apoE4 knock-in [11], activated endothelial cells [12], and microglia-derived fragmented mitochondria [13]. However, other factors/conditions induce neuroprotective and anti-inflammatory astrocytes, including reduction in P2Y1 receptor expression [14], ischemia-reperfusion [15], spinal cord injury [16], and type I interferons and microbial metabolite tryptophan [17,18]. These reports suggest that astrocytic neurodegenerative inflammatory responses or neuroprotective anti-inflammatory responses can be controlled by various factors, and that the inflammation caused by astrocytes would be a novel therapeutic target for neurodegenerative diseases. In this review, we discuss the pathogenic roles of astrocytes and Müller cells in glaucoma, especially focusing on their pro-inflammatory responses (hereafter referred to as “immune responses”).

2. Glaucoma

Glaucoma, a progressive optic neuropathy that affects more than 70 million people worldwide [19,20], is the second most common cause of blindness worldwide. In glaucoma, blindness is caused by damage to the optic nerve and degeneration of retinal ganglion cells (RGCs), retinal neurons essential for transducing visual information to the brain. Elevated intraocular pressure (IOP) is one of the most well-known risk factors for glaucoma, and the simplest interpretation is that IOP-mediated stress damages RGCs (Figure 1). Although reducing IOP can prevent or delay vision loss [21,22], IOP elevation alone does not determine whether patients develop glaucoma, and many patients develop glaucoma regardless of their IOP level [22]. In addition, some individuals show ocular hypertension, a diagnosis applied when the IOP is above the normal range, without any optic nerve damage [23]. Importantly, a substantial percentage of glaucoma patients (approximately 30%–40% in Caucasians) show normal IOP levels, i.e., normal-tension glaucoma (NTG) [24]. The Asian, and especially Japanese [25], population [26] shows a significantly higher prevalence of NTG (52%–92%). These reports suggest that risk factors other than IOP are also important for the pathogenesis of glaucoma.
Glaucomatous ocular tissue damage includes anatomical and functional deterioration of the retina and optic nerve head (ONH). The optic disc in the ONH is the location of the retina at which RGC axons gather together, form an optic nerve, and exit the eye [27,28]. The optic disc under ophthalmoscopy is observed as a blight circle near the center part of the eye. There is a bright spot at the center of the optic disc, namely the ‘(optic) cup’. Enlargement of ONH excavation (referred to as ‘cupping’, i.e., narrowing of the neuronal rim and an increase in the cup-to-disk ratio) is the hallmark of anatomical changes in human patients with hypertensive glaucoma (primary open-angle glaucoma, POAG) and NTG [27] (Figure 2). The ONH has been suggested to be the primary site of injury in glaucoma. The primate glaucoma model and post-mortem tissue of glaucoma patients revealed that RGC axons are lost at the lamina cribrosa [29,30]. The lamina cribrosa is an extracellular matrix (ECM)-rich (mainly collagen) structure, located just behind the optic cup, which provides physical support to the axon fibers as they pass through the posterior wall of the eye. This collagen-rich structure (i.e., ECM plate) is limited in the lamina cribrosa of humans and non-human primates. In rodents, since the region corresponding to the human LC lacks the ECM plate but astrocytes are abundant, this region is referred to as the “glial lamina” [31]. Optic nerve atrophy at the ONH precedes visual impairment in glaucoma [32], and Wallerian degeneration-like mechanisms may cause RGC soma death [33]. In addition to the RGC soma loss, thinning of the retinal nerve fiber layer, a well-known tissue change found in glaucoma patients, is associated with visual field loss [27]. Dendritic atrophy is also a characteristic feature of RGCs in both glaucoma patients [34] and model animals [35]. Such RGC damage and death induce visual impairment in glaucoma.
Elevated IOP is a well-known risk factor for hypertensive glaucoma (POAG). The simplest hypothesis is that IOP-mediated physical stress may directly damage retinal ganglion cells (RGCs) and the optic nerve. Accumulating evidence suggests that elevated IOP alone does not determine whether patients develop glaucoma but that other factors also contribute to the pathogenesis of glaucoma. In the case of NTG, glaucoma is developed even if the patients have a normal level of IOP.
In glaucoma patients, a central depression of the retina (i.e., the optic cup) is deformed, leading to posterior displacement and enlargement of the optic cup. The lamina cribrosa, a collagen-enriched tissue, provides physical support to the retinal ganglion cell (RGC) axons. With the enlargement of the optic cup, the structural remodeling of the lamina cribrosa physically stresses and damages the optic nerve. The normal and neuroprotective astrocytes support RGC axon integrity by releasing neurotrophic factors and/or anti-inflammatory molecules. Under glaucoma, astrocytes release neurotoxic factors such as cytokines and chemokines and damage RGC axons.

3. Astrocytes, Immune Responses, and Pathogenesis of Glaucoma

The retina, ONH, and optic nerve contain several glial types, such as astrocytes, Müller cells, and microglia (Figure 3). Astrocytes localize at the inner surface of the retina and throughout the optic nerve. Müller cells exist only in the retina and vertically span the entire thickness of the retina. Oligodendrocytes exist in the distal part of the optic nerve. GFAP-positive astrocytes have been shown to be closely associated with Tuj1-positive RGC axons in mouse retinal slices (Figure 4a). Visualization of Müller cells by YC-Nano under the control of Mlc-tTA [36] revealed that they extend their processes toward and attach to SMI32-positive RGC somas (asterisk, Figure 4b). Some Müller cell end feet enwrap RGC axons (arrow, Figure 4b). At the ONH, astrocytes are accumulated along with RGC axons (Figure 4c). Horizontal slices of ONH show that GFAP-positive astrocytes form honeycomb structures, and that SMI32-positive RGC axons pass through them (Figure 4d). Similar to those found in the brain, ocular astrocyte-lineage cells help maintain homeostatic retinal functions, including metabolic support and nutrition of neurons, ion buffering, water transport, neurotransmitter uptake, and blood vessel regulation, and they can act as optical fibers [37,38,39].
Inflammation may be related to the pathogenesis of glaucoma [40,41]. The complement pathway, essential for immune responses and inflammation, may also be related to the pathogenesis of glaucoma. Retinal astrocytes up-regulate complement C1q expression in RGCs [42]. The DBA/2J mouse, an inherited glaucoma model that develops elevated IOP and RGC death, shows up-regulated retinal C1q expression. Because C1q and its downstream C3 signaling mediate synapse elimination, overactivation of these signaling pathways may be an initial cause of the pathogenesis of glaucoma. Recent single-cell RNA-sequence analysis revealed that Müller cells are the major contributor to complement activations in the retina [43]. Aging, an important risk factor for glaucoma, increases various complement expressions in the retina. Transient ocular ischemia also up-regulates multiple complement molecules; this is not limited to rodents, but is also observed in the non-human primate model of glaucoma and human glaucoma patients [44]. These reports suggest that both astrocytes and Müller cells may contribute to evoking immune responses and the pathogenesis of glaucoma.
A schematic showing glial cells in the retina. In addition to retinal neurons (e.g., RGCs, ACs, BPs, HCs, and rod and cone photoreceptors), there are several types of glial cells in the retina. Astrocytes are located at the inner surface of the retina. Müller cells vertically span the entire thickness of the retina. Microglia exist in various neuronal layers, such as the NFL, GCL, IPL, and OPL. NFL: nerve fiber layer; GCL: ganglion cell layer; IPL: inner plexiform layer; INL: inner nuclear layer; OPL: outer plexiform layer; ONL: outer nuclear layer; IS/OS: inner or outer segment of photoreceptors; RGC: retinal ganglion cell; AC: amacrine cell; BP: bipolar cell; HC: horizontal cell.
Extracellular ATP may also regulate immune responses by astrocyte-lineage cells. Extracellular ATP binds to receptors in the plasma membrane, namely purinergic P2 receptors. ATP-mediated purinergic signaling is an important regulator for damage-associated molecular patterns or alarmins [45]. ATP is released or leaked from various cells and tissues in the eye [46]. Under physiological conditions, extracellular ATP levels are tightly regulated and maintained at very low concentrations. Under pathological conditions, extracellular ATP levels are often elevated by leakage and/or release from damaged cells or other cell types. High ATP concentrations activate P2X7 receptors, a high-threshold type of P2 receptor, and trigger pro-inflammatory responses via astrocyte-lineage cells. Activation of astrocyte P2X7 receptors induces the production of chemokines, cytokines, and reactive oxygen species, and the priming of the nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing protein 3, an essential inflammasome regulator [47,48,49]. P2X7 receptor activation also induces cytokine production in Müller cells [50]. Astrocyte-mediated immune responses may regulate neuronal functions and damage neurons via controlling microglia or peripheral immune cells [51,52,53,54]. Immune cell responses may regulate microglia-to-astrocyte signaling and neurotoxicity. Microglia can release ATP by exocytosis [55], and activation of microglial TLR4 triggers ATP release and P2Y1 receptor activation in astrocytes, which induces glutamate release and enhances excitatory neurotransmission [56]. Another study has shown that microglia-derived ATP activates the astrocytic P2Y1 receptor and induces interleukin (IL)-6 expression, thereby inducing a neuroprotective effect [57] by blocking excitatory neurotransmission [58]. Astrocytic or microglial CXCR4 signaling enhances astrocytic glutamate release and causes neurotoxicity [59]. A recent study has suggested that C-X-C motif chemokine ligand 12 (CXCL12), an endogenous agonist for CXCR4, is up-regulated in retinal astrocytes in an NTG mouse model [60].
Previous reports have suggested that microglia dynamically regulate neuroprotective [14] or neurotoxic [9] astrocytic phenotypes. Neurotoxic astrocytes are induced by tumor necrosis factor (TNF)/IL-1α/C1q signals from microglia, and blocking this signaling protects RGCs from optic nerve crush damage. TNFα is also released from astrocytes, and astrocyte-derived TNFα enhances glutamate-mediated excitatory neurotransmission [61]. Although the hypothesis of glutamate-mediated RGC excitotoxicity in glaucoma patients is still under debate [62], many animal models have suggested that excitotoxicity is an attractive candidate for glaucoma. A recent study has proposed that astrocytic immune responses may cause excitotoxicity of RGCs under glaucomatous conditions [60]. A novel NTG mouse model showed that astrocytic immune responses were associated with a reduction in Grin3a expression in RGCs. NR3A, a dominant-negative subunit of the Grin3a-encoding N-methyl-D-aspartic acid (NMDA) receptor [63], suppresses Ca2+ permeability through the NMDA channel. In the novel NTG mouse model, certain RGC subgroups expressed high Grin3a expression, which was dramatically decreased in glaucoma [60]. Grin3a deficiency causes Ca2+ overload and excitotoxicity in RGCs [63,64]. The immune functions of astrocyte-lineage cells may be involved in glutamate-mediated RGC toxicity in glaucoma.

4. Autoimmunity and Glaucoma

Over the past decades, accumulating evidence from clinical and basic studies has suggested that dysregulated immune systems and autoimmunity may trigger the pathogenesis of glaucoma. Astrocyte-lineage cells may bridge autoimmunity and glaucoma. Abnormal immune activity can be observed [65] in glaucoma patients, of which many show an elevated prevalence of monoclonal gammopathy [66]. Glaucoma patients also show elevated levels of autoantibodies against crystallin, S100, and heat shock proteins (HSPs) in tears, serum [67,68,69], and aqueous humor [70].
The gene encoding αB-crystallin (CRYAB) is highly expressed in astrocytes and Müller cells in the human retina [71]. CRYAB was initially identified as a negative regulator of T-cell responses in experimental autoimmune encephalomyelitis [72], whereas phosphorylation of intracellular CRYAB in astrocytes mediates reactive astrogliosis and worsens the pathology of multiple sclerosis in both the mouse and human brain [73]. In the mouse model of glaucoma, the protein expression of crystallin decreased with age, and recombinant crystallin proteins protected RGCs in retinal explants [74]. Although CRYAB in astrocytes plays detrimental roles in the brain, reduced CRYAB levels may be linked to higher T-cell responses. An animal model has proposed that T cells play an active role in the pathogenesis of glaucoma [75].
To reproduce glaucoma autoimmunity, researchers employed immunization of animals by ocular tissue antigen homogenate and found that the immunized animals showed increased levels of autoreactive antibodies against ocular tissue [76]. The immunized animals also exhibited glaucoma-like tissue changes, such as RGC death, optic nerve atrophy, and reduced ocular responses without elevated IOP [77]. In addition to the tissue homogenates, S100β and HSPs were used for immunization. S100β, a Ca2+-binding protein, is highly expressed in astrocytes and Müller cells, which release S100β in response to various stimuli such as neuronal activity and high glucose [78,79]. When S100β protein is intravitreally administered, RGCs degenerate without changes to IOP [80]. S100β increases the expression of NFκB and complement C3 [81] and triggers microglial pro-inflammatory responses [82,83]. The S100β-evoked RGC damage and impaired ocular responses are recovered by minocycline [84], indicating the role of microglia in triggering glaucoma-like phenotypes. Whether S100β immunization induces T-cell responses is currently unclear. Autoimmunity against HSPs may also be related to glaucoma, and serum anti-HSPs antibodies were elevated in serum [67,68,69] and aqueous humor [70] of glaucoma patients. Because ONH astrocytes express and up-regulate Hsp27 in glaucoma patients [85], they might release HSPs and participate in autoimmune responses and RGC damage. In the case of HSPs, T cells infiltrate and damage RGCs by releasing Fas-ligand [86].

5. Glaucoma Risk Genes and Astrocyte-Lineage Cells

A recent study revealed that astrocyte-derived neurotoxic molecules include long saturated fatty acids and ELOVL fatty acid elongase 1 (ELOVL1) [87]. Previous genome-wide association studies have suggested that single-nucleotide polymorphisms (SNPs) of the ELOVL5 gene correlate with a higher risk for NTG [88] and primary open-angle glaucoma [89]. Notably, the human protein atlas database (https://www.proteinatlas.org/, accessed on 28 January 2023) and previous single-cell transcriptome data from human retina have revealed that the ELOVL5 gene is highly enriched in Müller cells rather than astrocytes [71,90,91]. These reports suggest lipid signaling also contributes to immune responses and the pathogenesis of glaucoma via glial cells.
Other immune response-related genes may also participate in the pathogenesis of glaucoma. The SNPs of the genes encoding Toll-like receptor-2 (TLR2) and -4 (TLR4), key molecules for anti-infectious and inflammatory responses, also correlate with a higher risk for NTG [92,93,94]. Although TLRs are highly enriched in the microglia of the brain, human retinal astrocytes highly express the TLR4 gene [71]. The SNPs of the gene encoding Lysyl oxidase-like 1 (LOXL1), related to inflammation and fibrosis in the liver and lungs [95,96], also correlate with elevated risk for NTG [97,98]. Recent single-cell transcriptome studies have revealed that the LOXL1 gene is abundant in Müller cells and astrocytes in humans [71]. Caveolin genes are also related to an elevated risk for NTG [99]. Both caveolin-1 and -2 genes (CAV1 and CAV2) are highly enriched in astrocytes and Müller cells in the human retina [71]. Cav1 knockout reduces lipopolysaccharide-evoked production of pro-inflammatory cytokines (CCL2, CXCL1, IL-6, and IL-1β) but increases the infiltration of leukocytes [100]. Cav1 deficiency also reduces IL-6 receptor-mediated STAT3 activation and protects retinal neurons against sodium iodate [101], indicating that aberrant astrocyte/Müller cell immune responses may be involved in glaucomatous neurodegeneration.
Glial cell senescence may be linked to the pathogenesis of glaucoma. The SIX homeobox 6 (SIX6) gene, a senescence-related gene, is also a risk gene for glaucoma, including NTG [102,103]. The SIX6 gene is highly expressed in astrocytes and Müller cells in the human retina [71,90,91]. A previous study has demonstrated that SIX6 induces the expression of the senescence gene p16INK4 and mediates the pathogenesis of glaucoma [104]. In the neurodegenerative disease mouse model, senescent astrocytes and microglia-protected neurons were depleted [105]. Because senescent cells release a group of soluble molecules known as senescence-associated secretory phenotype factors (e.g., chemokines, cytokines, and growth factors), immune responses may participate in astrocyte senescence-mediated neuronal damage. Gene variation of SIX homeobox 1 (SIX1) is also related to a higher risk for glaucoma [103,106]. SIX1 also controls senescence via p16INK4 [107] and is exclusively expressed in Müller cells [71]. Because SIX1 is an integral component of the non-canonical component of NFκB activation and suppresses inflammation [108], a functional deficit of SIX1 in Müller cells could induce their inflammatory responses.
Intracellular Ca2+ is a well-known essential signal for regulating astrocyte functions [109,110]. Transmembrane and coiled-coil domains 1 is a Ca2+-load-activated Ca2+ channel that is expressed in and regulates Ca2+ levels within the endoplasmic reticulum [111]; its deficiency results in Ca2+ overflow into the endoplasmic reticulum [112]. Aberrant Ca2+ signaling in astrocytes is linked to neurological disease [113] and is often associated with pro-inflammatory responses.
Connexin 43 encoded by the Gja1 gene also has pivotal roles in maintaining the ocular microenvironment, and a recent study has suggested that GJA1 gene variation is associated with an elevated risk for glaucoma [114] because astrocytic connexin is essential for gap junction formation. The astrocytic gap junction contributes to buffering of K+ ions and glutamate incorporated from the synaptic cleft. Dysfunction in gap junction formation limits astrocyte-mediated clearance of K+ and glutamate and causes neuronal hyperexcitability, which may be related to neurodegenerative diseases [115]. Associated with excitotoxicity, deficiency in astrocyte Cx43 exacerbates inflammation after brain injury [116]. In human patients with glaucoma, GJA1 expression is higher than in age-matched healthy volunteers [117]. Because the Cx43 spatial pattern is changed in glaucoma [118], the gap junction may be lost under pathological conditions. High hydrostatic pressure causes internalization of Cx43 in human ONH astrocytes, indicating that the gap junction decouples intercellular communication under glaucomatous conditions [119]. The decoupled Cx43 (i.e., hemichannel) mediates ATP release and triggers inflammation. These reports suggest that many glaucoma-risk genes are expressed in astrocyte-lineage cells, and their dysfunctions may cause the pathogenesis and progression of glaucoma.

6. Astrocyte Responses in Glaucoma Patients

In glaucoma patients, ocular astrocyte-lineage cells show dynamic structural and molecular changes [30,120,121,122,123,124,125]. Similar to other species, human astrocytes are also present at the inner surface of the retina along with RGC axons. They intimately associate with RGC axons and are highly accumulated at the ONH and optic nerve. As described above, the lamina cribrosa, a mesh-like structure that allows axons of the optic nerve to pass through the sclera in the ONH, is the primary site of damage in RGC axons [126,127]. The intraorbital and retrolaminar optic nerve is unmyelinated and thus directly surrounded by astrocytes [128], indicating that dysfunction of astrocytes at this site is critical for the pathogenesis of glaucoma.
A previous study has shown that ONH astrocytes become reactive in glaucoma patients [126]. In an animal model of glaucoma, such changes are the earliest detectable event that is often observed before axonal damage [129]. As described above, ONH cupping is essential for axonal damage (Figure 2) and is induced by the reorganization of existing tissue via tissue destruction and ECM production. ONH astrocytes from glaucoma patients also express higher levels of matrix metalloproteinases (MMPs) [130] and ECM molecules [123]. Transforming growth factor β (TGFβ) mediates this process and is expressed in ONH astrocytes [131,132]. TGFβ levels in the ONH and aqueous humor are significantly higher in glaucoma patients [131]. TGFβ increases the expression levels of ECM and ECM-degrading enzymes, including MMPs [133]. Statin, a potent inhibitor of cholesterol synthesis, suppresses TGFβ-mediated MMP expression in human ONH astrocytes [134]. A recent study has suggested that deficiency in astrocytic ATP-binding cassette transporter A1, which transports cholesterol and fatty acids to the ECM [135], induces astrocytic pro-inflammatory responses and NTG-like pathology in mice [60]. Abnormal lipid signaling in astrocytes may accelerate tissue remodeling of the ONH, and a previous study has suggested that oxidative stress is involved in TGFβ-mediated ECM production in human ONH astrocytes [136]. Purinergic P2Y1 receptor signaling up-regulates antioxidant genes in astrocytes [137,138] and thus may be an attractive target for preventing ONH remodeling. In support of this hypothesis, P2Y1 receptor deficiency causes age-associated RGC damage [139]. Dysregulated extracellular ATP-mediated glial signaling can cause oxidative stress and damage RGCs in glaucoma [46].
In addition to tissue remodeling, inflammatory responses by astrocytes may be important for RGC damage. Human ONH astrocytes express nitric oxide synthase and up-regulate its expression in glaucoma [140,141], which may damage RGC axons [142,143]. ONH astrocytes from NTG patients also express TNFα [130]. In addition to astrocytes, Müller cells may contribute to RGC degeneration in glaucoma patients [144]. Up-regulated TNFα expression was observed in Müller cells in the retina of glaucoma patients [145]. Advanced glycation end-products are increased in glaucoma patients, and their receptors, essential regulators of inflammation [146], are up-regulated mainly in Müller cells in glaucoma patients [147]. Mice with advanced glycation end-product receptor knockout show a protective effect against ocular hypertension in RGCs [148], indicating that the inflammatory responses by Müller cells may cause RGC damage. At the perivascular region of the retina, reactive changes in astrocytes and Müller cells are observed in the early stages of glaucoma [149], and the retinal area at which astrocyte-lineage cells become reactive shows a significantly thinner retinal nerve fiber layer [150]. These studies suggest that immune responses of astrocyte-lineage cells are essential components of glaucoma pathogenesis.
Another molecule related to glaucoma is endothelin (ET), a potent vasoconstrictor. Plasma ET-1 levels are significantly elevated in NTG patients [151,152]. As intravitreal administration of ET-1 decreases IOP [151], ET-1-mediated RGC damage seems irrelevant for IOP elevation. Higher plasma ET-1 levels and SNPs of the genes encoding ET and ET receptors (ETRs) correlate with a higher risk for NTG [153,154,155,156]. Abnormal ET signaling could be involved in altered ocular circulation. Impaired ocular blood flow and disc hemorrhage are involved in the onset and progression of NTG [157,158,159]. In addition, immune responses by astrocyte-lineage cells contribute to the pathogenic roles of ET. The single-cell RNA-sequence database from the human retina has shown that ET and ETR are highly expressed in astrocytes and Müller cells [71,90,91]. Stimulation of human ONH astrocytes with ET-1 triggers [Ca2+]i transients and cell proliferation, which are mediated by ETAR and ETBR [160]. ET-2 induces reactive changes in Müller cells, disruption of the blood-retina barrier, and infiltration of peripheral immune cells [161]. The blockade of peripheral immune cells and treatment with bosentan, an ETR antagonist, show a protective effect on RGCs in DBA/2J mice [162,163]. ET-1 also increases the expression of MMP, tissue inhibitor metalloproteinase, and ECM in ONH astrocytes [164]. Taken together, immune responses by astrocyte-lineage cells are involved in the onset and progression of glaucoma.

7. Astrocytes as a Therapeutic Target for Glaucoma

As described above, previous studies have suggested that abnormal astrocytes trigger and accelerate the pathology of glaucoma. The prevention of long saturated fatty acids by knocking ELOVL1 out of astrocytes protected RGCs against optic nerve crush [87]. In the case of astrocytic ABCA1, the loss of ABCA1 causes cholesterol accumulation in astrocytes [60], which reminds us that the treatment of cholesterol-lowering drugs would be effective. In human patients with glaucoma, the use of statin (a cholesterol—lowering agent) is associated with a reduced risk of having glaucoma [165], suggesting that preventing cholesterol accumulation in astrocytes is also an attractive therapeutic target for glaucoma. Other studies have shown that connexin 43 (Cx43)-mediated astrocytic coupling is another therapeutic target for glaucoma [166]. Exposing human ONH astrocytes to high hydrostatic pressure causes internalization of Cx43 and decouples intercellular communication through gap junction [119]. Because connexin hemichannel is linked to inflammation, drugs maintaining the Cx43 gap junction (e.g., Dnegaptide) may be a candidate for astrocyte-targeted glaucoma treatment. A recent report has shown that overexpression of secreted phosphoprotein 1 (i.e., osteopontin) in astrocytes induces anti-inflammatory responses and protects RGCs [167]. In addition to blocking neurotoxic astrocytes, acceleration of neuroprotective astrocyte function may be an attractive candidate for glaucoma treatment.

8. Concluding Remarks

Astrocytes and tissue-specific astrocyte-lineage cells are widely distributed in the nervous system. Ocular astrocyte-lineage cells are essential for maintaining the physiological microenvironment around retinal neurons and optic nerves. Reactive changes and dysfunctions of these cell types are often found in human patients with ocular diseases such as glaucoma. At least a part of the pathological changes in glaucoma patients seems to be mediated by them. Many glaucoma-related molecules are expressed in astrocyte-lineage cells and cause their immune responses (Figure 5). As accumulating evidence suggests pathogenic roles of astrocytes in various neurodegenerative diseases, ocular astrocyte-lineage cells might be a novel therapeutic target for glaucoma in the near future.
Transforming growth factor (TGFβ) and endothelin (ET-1) levels are elevated in serum and/or aqueous humor of glaucoma patients. These molecules up-regulate the expression of matrix metalloproteinases (MMPs) and extracellular matrix (ECM) proteins in the optic nerve head (ONH), which causes tissue remodeling. Purinergic signaling may be altered in glaucoma. Excess amounts of ATP could activate P2X7 receptors and induce inflammation, whereas activation of the P2Y1 receptor induces antioxidative responses that may suppress ONH remodeling. ONH astrocytes from glaucoma patients also show higher production of nitric oxide (NO) and tumor necrosis factor (TNFα), which may damage retinal ganglion cells (RGCs). Advanced glycation end-products (AGEs) are also increased in glaucoma patients’ eyes and can activate advanced glycation end-product receptors (RAGE) expressed in astrocyte-lineage cells and trigger inflammation. Many glaucoma risk genes are expressed in both astrocytes and Müller cells and contribute to their inflammatory responses. Molecules highly enriched in astrocyte-lineage cells may activate microglia and T cells to induce autoimmune glaucoma.

Author Contributions

Original draft preparation, Y.S.; review and editing, K.K. and S.K.; supervision, K.K. and S.K.; funding acquisition, Y.S. and K.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Takeda Science Foundation (Y.S. and S.K.); Mitsubishi Foundation (S.K.); Japan Agency for Medical Research and Development (grant number 20gm1310008s0101 to S.K.); Japan Society for the Promotion of Science KAKENHI grant numbers JP16K18390 (Y.S.), JP18K06481 (Y.S.), JP20KK0366 (Y.S.), JP16H04669 (S.K.), JP25117003 (S.K.), JP18H05121 (S.K.), JP19H04746 (S.K.), JP20H05902 (S.K.), JP20H05984 (S.K.), and JP21H04786 (S.K.); and a Grant-in-Aid for Scientific Research on Innovative Areas-Resource and Technical Support Platforms for Promoting Research “Advanced Bioimaging Support” (JP16H06280) (Y.S.). This study was also partially supported by JST Grant CREST number JPMJCR14G2 (S.K.), AMED-CREST grant number JP20gm131000 (S.K.), and a Frontier Project Grant from the University of Yamanashi (S.K.). This research was also supported by Joint Research of the Exploratory Research Center on Life and Living Systems (ExCELLS; program number 21-414) (Y.S.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Eva Lasic from Edanz Group (https://en-author-services.edanz.com/, accessed on 27 January 2023) for editing a draft of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Allen, N.J.; Barres, B.A. Neuroscience: Glia—More than Just Brain Glue. Nature 2009, 457, 675–677. [Google Scholar] [CrossRef] [PubMed]
  2. Brandebura, A.N.; Paumier, A.; Onur, T.S.; Allen, N.J. Astrocyte Contribution to Dysfunction, Risk and Progression in Neurodegenerative Disorders. Nat. Rev. Neurosci. 2022, 24, 23–39. [Google Scholar] [CrossRef] [PubMed]
  3. Miller, G. Neuroscience. The Dark Side of Glia. Science 2005, 308, 778–781. [Google Scholar] [CrossRef]
  4. Habib, N.; McCabe, C.; Medina, S.; Varshavsky, M.; Kitsberg, D.; Dvir-Szternfeld, R.; Green, G.; Dionne, D.; Nguyen, L.; Marshall, J.L.; et al. Disease-Associated Astrocytes in Alzheimer’s Disease and Aging. Nat. Neurosci. 2020, 23, 701–706. [Google Scholar] [CrossRef] [PubMed]
  5. Gu, X.-L.; Long, C.-X.; Sun, L.; Xie, C.; Lin, X.; Cai, H. Astrocytic Expression of Parkinson’s Disease-Related A53T Alpha-Synuclein Causes Neurodegeneration in Mice. Mol. Brain 2010, 3, 12. [Google Scholar] [CrossRef]
  6. Tong, X.; Ao, Y.; Faas, G.C.; Nwaobi, S.E.; Xu, J.; Haustein, M.D.; Anderson, M.A.; Mody, I.; Olsen, M.L.; Sofroniew, M.V.; et al. Astrocyte Kir4.1 Ion Channel Deficits Contribute to Neuronal Dysfunction in Huntington’s Disease Model Mice. Nat. Neurosci. 2014, 17, 694–703. [Google Scholar] [CrossRef] [PubMed]
  7. Yamanaka, K.; Chun, S.J.; Boillee, S.; Fujimori-Tonou, N.; Yamashita, H.; Gutmann, D.H.; Takahashi, R.; Misawa, H.; Cleveland, D.W. Astrocytes as Determinants of Disease Progression in Inherited Amyotrophic Lateral Sclerosis. Nat. Neurosci. 2008, 11, 251–253. [Google Scholar] [CrossRef] [PubMed]
  8. Wheeler, M.A.; Clark, I.C.; Tjon, E.C.; Li, Z.; Zandee, S.E.J.; Couturier, C.P.; Watson, B.R.; Scalisi, G.; Alkwai, S.; Rothhammer, V.; et al. MAFG-Driven Astrocytes Promote CNS Inflammation. Nature 2020, 578, 593–599. [Google Scholar] [CrossRef]
  9. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C.; et al. Neurotoxic Reactive Astrocytes Are Induced by Activated Microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
  10. Clarke, L.E.; Liddelow, S.A.; Chakraborty, C.; Münch, A.E.; Heiman, M.; Barres, B.A. Normal Aging Induces A1-like Astrocyte Reactivity. Proc. Natl. Acad. Sci. USA 2018, 115, E1896–E1905. [Google Scholar] [CrossRef] [Green Version]
  11. Shi, Y.; Yamada, K.; Liddelow, S.A.; Smith, S.T.; Zhao, L.; Luo, W.; Tsai, R.M.; Spina, S.; Grinberg, L.T.; Rojas, J.C.; et al. ApoE4 Markedly Exacerbates Tau-Mediated Neurodegeneration in a Mouse Model of Tauopathy. Nature 2017, 549, 523–527. [Google Scholar] [CrossRef] [PubMed]
  12. Taylor, X.; Cisternas, P.; Jury, N.; Martinez, P.; Huang, X.; You, Y.; Redding-Ochoa, J.; Vidal, R.; Zhang, J.; Troncoso, J.; et al. Activated Endothelial Cells Induce a Distinct Type of Astrocytic Reactivity. Commun Biol 2022, 5, 282. [Google Scholar] [CrossRef]
  13. Joshi, A.U.; Minhas, P.S.; Liddelow, S.A.; Haileselassie, B.; Andreasson, K.I.; Dorn, G.W., 2nd; Mochly-Rosen, D. Fragmented Mitochondria Released from Microglia Trigger A1 Astrocytic Response and Propagate Inflammatory Neurodegeneration. Nat. Neurosci. 2019, 22, 1635–1648. [Google Scholar] [CrossRef] [PubMed]
  14. Shinozaki, Y.; Shibata, K.; Yoshida, K.; Shigetomi, E.; Gachet, C.; Ikenaka, K.; Tanaka, K.F.; Koizumi, S. Transformation of Astrocytes to a Neuroprotective Phenotype by Microglia via P2Y1 Receptor Downregulation. Cell Rep. 2017, 19, 1151–1164. [Google Scholar] [CrossRef]
  15. Zamanian, J.L.; Xu, L.; Foo, L.C.; Nouri, N.; Zhou, L.; Giffard, R.G.; Barres, B.A. Genomic Analysis of Reactive Astrogliosis. J. Neurosci. 2012, 32, 6391–6410. [Google Scholar] [CrossRef] [PubMed]
  16. Faulkner, J.R.; Herrmann, J.E.; Woo, M.J.; Tansey, K.E.; Doan, N.B.; Sofroniew, M.V. Reactive Astrocytes Protect Tissue and Preserve Function after Spinal Cord Injury. J. Neurosci. 2004, 24, 2143–2155. [Google Scholar] [CrossRef] [PubMed]
  17. Rothhammer, V.; Mascanfroni, I.D.; Bunse, L.; Takenaka, M.C.; Kenison, J.E.; Mayo, L.; Chao, C.-C.; Patel, B.; Yan, R.; Blain, M.; et al. Type I Interferons and Microbial Metabolites of Tryptophan Modulate Astrocyte Activity and Central Nervous System Inflammation via the Aryl Hydrocarbon Receptor. Nat. Med. 2016, 22, 586–597. [Google Scholar] [CrossRef] [PubMed]
  18. Rothhammer, V.; Borucki, D.M.; Tjon, E.C.; Takenaka, M.C.; Chao, C.-C.; Ardura-Fabregat, A.; de Lima, K.A.; Gutiérrez-Vázquez, C.; Hewson, P.; Staszewski, O.; et al. Microglial Control of Astrocytes in Response to Microbial Metabolites. Nature 2018, 557, 724–728. [Google Scholar] [CrossRef] [PubMed]
  19. Weinreb, R.N.; Aung, T.; Medeiros, F.A. The Pathophysiology and Treatment of Glaucoma: A Review. JAMA 2014, 311, 1901–1911. [Google Scholar] [CrossRef] [PubMed]
  20. Quigley, H.A. Number of People with Glaucoma Worldwide. Br. J. Ophthalmol. 1996, 80, 389–393. [Google Scholar] [CrossRef] [Green Version]
  21. Heijl, A.; Leske, M.C.; Bengtsson, B.; Hyman, L.; Bengtsson, B.; Hussein, M. Early Manifest Glaucoma Trial Group Reduction of Intraocular Pressure and Glaucoma Progression: Results from the Early Manifest Glaucoma Trial. Arch. Ophthalmol. 2002, 120, 1268–1279. [Google Scholar] [CrossRef]
  22. Kass, M.A.; Heuer, D.K.; Higginbotham, E.J.; Johnson, C.A.; Keltner, J.L.; Miller, J.P.; Parrish, R.K., 2nd; Wilson, M.R.; Gordon, M.O. 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. 2002, 120, 701–713, discussion 829–830. [Google Scholar] [CrossRef]
  23. Comparison of Glaucomatous Progression between Untreated Patients with Normal-Tension Glaucoma and Patients with Therapeutically Reduced Intraocular Pressures. Am. J. Ophthalmol. 1998, 126, 487–497. [CrossRef] [PubMed]
  24. Bonomi, L.; Marchini, G.; Marraffa, M.; Bernardi, P.; De Franco, I.; Perfetti, S.; Varotto, A.; Tenna, V. Prevalence of Glaucoma and Intraocular Pressure Distribution in a Defined Population. The Egna-Neumarkt Study. Ophthalmology 1998, 105, 209–215. [Google Scholar] [CrossRef]
  25. Iwase, A.; Suzuki, Y.; Araie, M.; Yamamoto, T.; Abe, H.; Shirato, S.; Kuwayama, Y.; Mishima, H.K.; Shimizu, H.; Tomita, G.; et al. The Prevalence of Primary Open-Angle Glaucoma in Japanese: The Tajimi Study. Ophthalmology 2004, 111, 1641–1648. [Google Scholar] [CrossRef] [PubMed]
  26. Cho, H.-K.; Kee, C. Population-Based Glaucoma Prevalence Studies in Asians. Surv. Ophthalmol. 2014, 59, 434–447. [Google Scholar] [CrossRef] [PubMed]
  27. Weinreb, R.N.; Leung, C.K.S.; Crowston, J.G.; Medeiros, F.A.; Friedman, D.S.; Wiggs, J.L.; Martin, K.R. Primary Open-Angle Glaucoma. Nat. Rev. Dis. Primers 2016, 2, 16067. [Google Scholar] [CrossRef]
  28. Shinozaki, Y.; Koizumi, S. Potential Roles of Astrocytes and Müller Cells in the Pathogenesis of Glaucoma. J. Pharmacol. Sci. 2021, 145, 262–267. [Google Scholar] [CrossRef] [PubMed]
  29. Burgoyne, C. The Morphological Difference between Glaucoma and Other Optic Neuropathies. J. Neuroophthalmol. 2015, 35 (Suppl. S1), S8–S21. [Google Scholar] [CrossRef] [PubMed]
  30. Quigley, H.A.; Addicks, E.M. Chronic Experimental Glaucoma in Primates. II. Effect of Extended Intraocular Pressure Elevation on Optic Nerve Head and Axonal Transport. Investig. Ophthalmol. Vis. Sci. 1980, 19, 137–152. [Google Scholar]
  31. Howell, G.R.; Libby, R.T.; Jakobs, T.C.; Smith, R.S.; Phalan, F.C.; Barter, J.W.; Barbay, J.M.; Marchant, J.K.; Mahesh, N.; Porciatti, V.; et al. Axons of Retinal Ganglion Cells Are Insulted in the Optic Nerve Early in DBA/2J Glaucoma. J. Cell Biol. 2007, 179, 1523–1537. [Google Scholar] [CrossRef] [Green Version]
  32. Sommer, A.; Katz, J.; Quigley, H.A.; Miller, N.R.; Robin, A.L.; Richter, R.C.; Witt, K.A. Clinically Detectable Nerve Fiber Atrophy Precedes the Onset of Glaucomatous Field Loss. Arch. Ophthalmol. 1991, 109, 77–83. [Google Scholar] [CrossRef] [PubMed]
  33. Howell, G.R.; Soto, I.; Libby, R.T.; John, S.W.M. Intrinsic Axonal Degeneration Pathways Are Critical for Glaucomatous Damage. Exp. Neurol. 2013, 246, 54–61. [Google Scholar] [CrossRef] [PubMed]
  34. Tribble, J.R.; Vasalauskaite, A.; Redmond, T.; Young, R.D.; Hassan, S.; Fautsch, M.P.; Sengpiel, F.; Williams, P.A.; Morgan, J.E. Midget Retinal Ganglion Cell Dendritic and Mitochondrial Degeneration Is an Early Feature of Human Glaucoma. Brain Commun 2019, 1, fcz035. [Google Scholar] [CrossRef] [PubMed]
  35. Harder, J.M.; Braine, C.E.; Williams, P.A.; Zhu, X.; MacNicoll, K.H.; Sousa, G.L.; Buchanan, R.A.; Smith, R.S.; Libby, R.T.; Howell, G.R.; et al. Early Immune Responses Are Independent of RGC Dysfunction in Glaucoma with Complement Component C3 Being Protective. Proc. Natl. Acad. Sci. USA 2017, 114, E3839–E3848. [Google Scholar] [CrossRef] [PubMed]
  36. Danjo, Y.; Shinozaki, Y.; Natsubori, A.; Kubota, Y.; Kashiwagi, K.; Tanaka, K.F.; Koizumi, S. The Mlc1 Promoter Directs Müller Cell-Specific Gene Expression in the Retina. Transl. Vis. Sci. Technol. 2022, 11, 25. [Google Scholar] [CrossRef] [PubMed]
  37. Vecino, E.; Rodriguez, F.D.; Ruzafa, N.; Pereiro, X.; Sharma, S.C. Glia-Neuron Interactions in the Mammalian Retina. Prog. Retin. Eye Res. 2016, 51, 1–40. [Google Scholar] [CrossRef] [PubMed]
  38. Newman, E.A. Glial Cell Regulation of Neuronal Activity and Blood Flow in the Retina by Release of Gliotransmitters. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2015, 370, 20140195. [Google Scholar] [CrossRef]
  39. Reichenbach, A.; Bringmann, A. New Functions of Müller Cells. Glia 2013, 61, 651–678. [Google Scholar] [CrossRef]
  40. Baudouin, C.; Kolko, M.; Melik-Parsadaniantz, S.; Messmer, E.M. Inflammation in Glaucoma: From the Back to the Front of the Eye, and Beyond. Prog. Retin. Eye Res. 2021, 83, 100916. [Google Scholar] [CrossRef]
  41. Tezel, G. Molecular Regulation of Neuroinflammation in Glaucoma: Current Knowledge and the Ongoing Search for New Treatment Targets. Prog. Retin. Eye Res. 2022, 87, 100998. [Google Scholar] [CrossRef] [PubMed]
  42. Stevens, B.; Allen, N.J.; Vazquez, L.E.; Howell, G.R.; Christopherson, K.S.; Nouri, N.; Micheva, K.D.; Mehalow, A.K.; Huberman, A.D.; Stafford, B.; et al. The Classical Complement Cascade Mediates CNS Synapse Elimination. Cell 2007, 131, 1164–1178. [Google Scholar] [CrossRef] [PubMed]
  43. Pauly, D.; Agarwal, D.; Dana, N.; Schäfer, N.; Biber, J.; Wunderlich, K.A.; Jabri, Y.; Straub, T.; Zhang, N.R.; Gautam, A.K.; et al. Cell-Type-Specific Complement Expression in the Healthy and Diseased Retina. Cell Rep. 2019, 29, 2835–2848.e4. [Google Scholar] [CrossRef]
  44. Stasi, K.; Nagel, D.; Yang, X.; Wang, R.-F.; Ren, L.; Podos, S.M.; Mittag, T.; Danias, J. Complement Component 1Q (C1Q) Upregulation in Retina of Murine, Primate, and Human Glaucomatous Eyes. Investig. Ophthalmol. Vis. Sci. 2006, 47, 1024–1029. [Google Scholar] [CrossRef]
  45. Di Virgilio, F.; Sarti, A.C.; Coutinho-Silva, R. Purinergic Signaling, DAMPs, and Inflammation. Am. J. Physiol. Cell Physiol. 2020, 318, C832–C835. [Google Scholar] [CrossRef]
  46. Shinozaki, Y.; Saito, K.; Kashiwagi, K.; Koizumi, S. Ocular P2 Receptors and Glaucoma. Neuropharmacology 2023, 222, 109302. [Google Scholar] [CrossRef] [PubMed]
  47. Panenka, W.; Jijon, H.; Herx, L.M.; Armstrong, J.N.; Feighan, D.; Wei, T.; Yong, V.W.; Ransohoff, R.M.; MacVicar, B.A. P2X7-like Receptor Activation in Astrocytes Increases Chemokine Monocyte Chemoattractant Protein-1 Expression via Mitogen-Activated Protein Kinase. J. Neurosci. 2001, 21, 7135–7142. [Google Scholar] [CrossRef]
  48. Munoz, F.M.; Patel, P.A.; Gao, X.; Mei, Y.; Xia, J.; Gilels, S.; Hu, H. Reactive Oxygen Species Play a Role in P2X7 Receptor-Mediated IL-6 Production in Spinal Astrocytes. Purinergic Signal. 2020, 16, 97–107. [Google Scholar] [CrossRef]
  49. Mitchell, C.H.; Albalawi, F.; Lim, J.; Lu, W. Priming of the NLRP3 Inflammasome in Optic Nerve Head Astrocytes by Mechanical Strain and Stimulation of the P2X7 Receptor. Investig. Ophthalmol. Vis. Sci. 2016, 57, 3009. [Google Scholar]
  50. Pelegrín, P. Many Ways to Dilate the P2X7 Receptor Pore. Br. J. Pharmacol. 2011, 163, 908–911. [Google Scholar] [CrossRef]
  51. McAlpine, C.S.; Park, J.; Griciuc, A.; Kim, E.; Choi, S.H.; Iwamoto, Y.; Kiss, M.G.; Christie, K.A.; Vinegoni, C.; Poller, W.C.; et al. Astrocytic Interleukin-3 Programs Microglia and Limits Alzheimer’s Disease. Nature 2021, 595, 701–706. [Google Scholar] [CrossRef]
  52. Vainchtein, I.D.; Chin, G.; Cho, F.S.; Kelley, K.W.; Miller, J.G.; Chien, E.C.; Liddelow, S.A.; Nguyen, P.T.; Nakao-Inoue, H.; Dorman, L.C.; et al. Astrocyte-Derived Interleukin-33 Promotes Microglial Synapse Engulfment and Neural Circuit Development. Science 2018, 359, 1269–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Endo, F.; Komine, O.; Fujimori-Tonou, N.; Katsuno, M.; Jin, S.; Watanabe, S.; Sobue, G.; Dezawa, M.; Wyss-Coray, T.; Yamanaka, K. Astrocyte-Derived TGF-Β1 Accelerates Disease Progression in ALS Mice by Interfering with the Neuroprotective Functions of Microglia and T Cells. Cell Rep. 2015, 11, 592–604. [Google Scholar] [CrossRef] [PubMed]
  54. Li, M.; Li, Z.; Yao, Y.; Jin, W.-N.; Wood, K.; Liu, Q.; Shi, F.-D.; Hao, J. Astrocyte-Derived Interleukin-15 Exacerbates Ischemic Brain Injury via Propagation of Cellular Immunity. Proc. Natl. Acad. Sci. USA 2017, 114, E396–E405. [Google Scholar] [CrossRef]
  55. Imura, Y.; Morizawa, Y.; Komatsu, R.; Shibata, K.; Shinozaki, Y.; Kasai, H.; Moriishi, K.; Moriyama, Y.; Koizumi, S. Microglia Release ATP by Exocytosis. Glia 2013, 61, 1320–1330. [Google Scholar] [CrossRef] [PubMed]
  56. Pascual, O.; Achour, S.B.; Rostaing, P.; Triller, A.; Bessis, A. Microglia Activation Triggers Astrocyte-Mediated Modulation of Excitatory Neurotransmission. Proc. Natl. Acad. Sci. USA 2012, 109, E197–E205. [Google Scholar] [CrossRef] [PubMed]
  57. Shinozaki, Y.; Nomura, M.; Iwatsuki, K.; Moriyama, Y.; Gachet, C.; Koizumi, S. Microglia Trigger Astrocyte-Mediated Neuroprotection via Purinergic Gliotransmission. Sci. Rep. 2014, 4, 4329. [Google Scholar] [CrossRef] [PubMed]
  58. Noguchi, Y.; Shinozaki, Y.; Fujishita, K.; Shibata, K.; Imura, Y.; Morizawa, Y.; Gachet, C.; Koizumi, S. Astrocytes Protect Neurons against Methylmercury via ATP/P2Y(1) Receptor-Mediated Pathways in Astrocytes. PLoS ONE 2013, 8, e57898. [Google Scholar] [CrossRef]
  59. Bezzi, P.; Domercq, M.; Brambilla, L.; Galli, R.; Schols, D.; De Clercq, E.; Vescovi, A.; Bagetta, G.; Kollias, G.; Meldolesi, J.; et al. CXCR4-Activated Astrocyte Glutamate Release via TNFalpha: Amplification by Microglia Triggers Neurotoxicity. Nat. Neurosci. 2001, 4, 702–710. [Google Scholar] [CrossRef] [PubMed]
  60. Shinozaki, Y.; Leung, A.; Namekata, K.; Saitoh, S.; Nguyen, H.B.; Takeda, A.; Danjo, Y.; Morizawa, Y.M.; Shigetomi, E.; Sano, F.; et al. Astrocytic Dysfunction Induced by ABCA1 Deficiency Causes Optic Neuropathy. Sci. Adv. 2022, 8, eabq1081. [Google Scholar] [CrossRef]
  61. Santello, M.; Bezzi, P.; Volterra, A. TNFα Controls Glutamatergic Gliotransmission in the Hippocampal Dentate Gyrus. Neuron 2011, 69, 988–1001. [Google Scholar] [CrossRef] [PubMed]
  62. Lotery, A.J. Glutamate Excitotoxicity in Glaucoma: Truth or Fiction? Eye 2005, 19, 369–370. [Google Scholar] [CrossRef] [Green Version]
  63. Sucher, N.J.; Kohler, K.; Tenneti, L.; Wong, H.-K.; Gründer, T.; Fauser, S.; Wheeler-Schilling, T.; Nakanishi, N.; Lipton, S.A.; Guenther, E. N-Methyl-D-Aspartate Receptor Subunit NR3A in the Retina: Developmental Expression, Cellular Localization, and Functional Aspects. Investig. Ophthalmol. Vis. Sci. 2003, 44, 4451–4456. [Google Scholar] [CrossRef] [PubMed]
  64. Nakanishi, N.; Tu, S.; Shin, Y.; Cui, J.; Kurokawa, T.; Zhang, D.; Chen, H.-S.V.; Tong, G.; Lipton, S.A. Neuroprotection by the NR3A Subunit of the NMDA Receptor. J. Neurosci. 2009, 29, 5260–5265. [Google Scholar] [CrossRef]
  65. Tezel, G.; Wax, M.B. Glaucoma. Chem. Immunol. Allergy 2007, 92, 221–227. [Google Scholar]
  66. Wax, M.B.; Tezel, G.; Edward, P.D. Clinical and Ocular Histopathological Findings in a Patient with Normal-Pressure Glaucoma. Arch. Ophthalmol. 1998, 116, 993–1001. [Google Scholar] [CrossRef] [PubMed]
  67. Grus, F.H.; Boehm, N.; Beck, S.; Schlich, M. Autoantibody Profiles in Tear Fluid as a Diagnostic Tool in Glaucoma. Investig. Ophthalmol. Vis. Sci. 2010, 51, 6110. [Google Scholar]
  68. Tezel, G.; Seigel, G.M.; Wax, M.B. Autoantibodies to Small Heat Shock Proteins in Glaucoma. Investig. Ophthalmol. Vis. Sci. 1998, 39, 2277–2287. [Google Scholar]
  69. Wax, M.B.; Tezel, G.; Kawase, K.; Kitazawa, Y. Serum Autoantibodies to Heat Shock Proteins in Glaucoma Patients from Japan and the United States. Ophthalmology 2001, 108, 296–302. [Google Scholar] [CrossRef]
  70. Joachim, S.C.; Bruns, K.; Lackner, K.J.; Pfeiffer, N.; Grus, F.H. Antibodies to Alpha B-Crystallin, Vimentin, and Heat Shock Protein 70 in Aqueous Humor of Patients with Normal Tension Glaucoma and IgG Antibody Patterns against Retinal Antigen in Aqueous Humor. Curr. Eye Res. 2007, 32, 501–509. [Google Scholar] [CrossRef]
  71. Lukowski, S.W.; Lo, C.Y.; Sharov, A.A.; Nguyen, Q.; Fang, L.; Hung, S.S.; Zhu, L.; Zhang, T.; Grünert, U.; Nguyen, T.; et al. A Single-Cell Transcriptome Atlas of the Adult Human Retina. EMBO J. 2019, 38, e100811. [Google Scholar] [CrossRef] [PubMed]
  72. Ousman, S.S.; Tomooka, B.H.; van Noort, J.M.; Wawrousek, E.F.; O’Connor, K.C.; Hafler, D.A.; Sobel, R.A.; Robinson, W.H.; Steinman, L. Protective and Therapeutic Role for AlphaB-Crystallin in Autoimmune Demyelination. Nature 2007, 448, 474–479. [Google Scholar] [CrossRef] [PubMed]
  73. Kuipers, H.F.; Yoon, J.; van Horssen, J.; Han, M.H.; Bollyky, P.L.; Palmer, T.D.; Steinman, L. Phosphorylation of AB-Crystallin Supports Reactive Astrogliosis in Demyelination. Proc. Natl. Acad. Sci. USA 2017, 114, E1745. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, H.; Bell, K.; Herrmann, A.; Arnhold, S.; Mercieca, K.; Anders, F.; Nagel-Wolfrum, K.; Thanos, S.; Prokosch, V. Crystallins Play a Crucial Role in Glaucoma and Promote Neuronal Cell Survival in an In Vitro Model Through Modulating Müller Cell Secretion. Investig. Ophthalmol. Vis. Sci. 2022, 63, 3. [Google Scholar] [CrossRef]
  75. Chen, H.; Cho, K.-S.; Vu, T.H.K.; Shen, C.-H.; Kaur, M.; Chen, G.; Mathew, R.; McHam, M.L.; Fazelat, A.; Lashkari, K.; et al. Commensal Microflora-Induced T Cell Responses Mediate Progressive Neurodegeneration in Glaucoma. Nat. Commun. 2018, 9, 3209. [Google Scholar] [CrossRef] [Green Version]
  76. Laspas, P.; Gramlich, O.W.; Müller, H.D.; Cuny, C.S.; Gottschling, P.F.; Pfeiffer, N.; Dick, H.B.; Joachim, S.C.; Grus, F.H. Autoreactive Antibodies and Loss of Retinal Ganglion Cells in Rats Induced by Immunization with Ocular Antigens. Investig. Ophthalmol. Vis. Sci. 2011, 52, 8835–8848. [Google Scholar] [CrossRef]
  77. Kuehn, S.; Meißner, W.; Grotegut, P.; Theiss, C.; Dick, H.B.; Joachim, S.C. Intravitreal S100B Injection Leads to Progressive Glaucoma Like Damage in Retina and Optic Nerve. Front. Cell. Neurosci. 2018, 12, 312. [Google Scholar] [CrossRef]
  78. Zong, H.; Ward, M.; Madden, A.; Yong, P.H.; Limb, G.A.; Curtis, T.M.; Stitt, A.W. Hyperglycaemia-Induced pro-Inflammatory Responses by Retinal Müller Glia Are Regulated by the Receptor for Advanced Glycation End-Products (RAGE). Diabetologia 2010, 53, 2656–2666. [Google Scholar] [CrossRef]
  79. Sakatani, S.; Seto-Ohshima, A.; Shinohara, Y.; Yamamoto, Y.; Yamamoto, H.; Itohara, S.; Hirase, H. Neural-Activity-Dependent Release of S100B from Astrocytes Enhances Kainate-Induced Gamma Oscillations in Vivo. J. Neurosci. 2008, 28, 10928–10936. [Google Scholar] [CrossRef]
  80. Tsai, T.; Joachim, S.C. Glaucoma-like Damage Induced by S100B Injection Is Accompanied by Microglial Response. Neural Regeneration Res. 2022, 17, 572–574. [Google Scholar]
  81. Reinehr, S.; Reinhard, J.; Gandej, M.; Gottschalk, I.; Stute, G.; Faissner, A.; Dick, H.B.; Joachim, S.C. S100B Immunization Triggers NFκB and Complement Activation in an Autoimmune Glaucoma Model. Sci. Rep. 2018, 8, 9821. [Google Scholar] [CrossRef] [PubMed]
  82. Bianchi, R.; Kastrisianaki, E.; Giambanco, I.; Donato, R. S100B Protein Stimulates Microglia Migration via RAGE-Dependent up-Regulation of Chemokine Expression and Release. J. Biol. Chem. 2011, 286, 7214–7226. [Google Scholar] [CrossRef] [PubMed]
  83. Grotegut, P.; Kuehn, S.; Meißner, W.; Dick, H.B.; Joachim, S.C. Intravitreal S100B Injection Triggers a Time-Dependent Microglia Response in a Pro-Inflammatory Manner in Retina and Optic Nerve. Mol. Neurobiol. 2020, 57, 1186–1202. [Google Scholar] [CrossRef] [PubMed]
  84. Grotegut, P.; Perumal, N.; Kuehn, S.; Smit, A.; Dick, H.B.; Grus, F.H.; Joachim, S.C. Minocycline Reduces Inflammatory Response and Cell Death in a S100B Retina Degeneration Model. J. Neuroinflammation 2020, 17, 375. [Google Scholar] [CrossRef] [PubMed]
  85. Tezel, G.; Hernandez, R.; Wax, M.B. Immunostaining of Heat Shock Proteins in the Retina and Optic Nerve Head of Normal and Glaucomatous Eyes. Arch. Ophthalmol. 2000, 118, 511–518. [Google Scholar] [CrossRef] [PubMed]
  86. Wax, M.B.; Tezel, G.; Yang, J.; Peng, G.; Patil, R.V.; Agarwal, N.; Sappington, R.M.; Calkins, D.J. Induced Autoimmunity to Heat Shock Proteins Elicits Glaucomatous Loss of Retinal Ganglion Cell Neurons via Activated T-Cell-Derived Fas-Ligand. J. Neurosci. 2008, 28, 12085–12096. [Google Scholar] [CrossRef] [Green Version]
  87. Guttenplan, K.A.; Weigel, M.K.; Prakash, P.; Wijewardhane, P.R.; Hasel, P.; Rufen-Blanchette, U.; Münch, A.E.; Blum, J.A.; Fine, J.; Neal, M.C.; et al. Neurotoxic Reactive Astrocytes Induce Cell Death via Saturated Lipids. Nature 2021, 599, 102–107. [Google Scholar] [CrossRef]
  88. Writing Committee for the Normal Tension Glaucoma Genetic Study Group of Japan Glaucoma Society; Meguro, A.; Inoko, H.; Ota, M.; Mizuki, N.; Bahram, S. Genome-Wide Association Study of Normal Tension Glaucoma: Common Variants in SRBD1 and ELOVL5 Contribute to Disease Susceptibility. Ophthalmology 2010, 117, 1331–1338.e5. [Google Scholar]
  89. Mabuchi, F.; Sakurada, Y.; Kashiwagi, K.; Yamagata, Z.; Iijima, H.; Tsukahara, S. Association between SRBD1 and ELOVL5 Gene Polymorphisms and Primary Open-Angle Glaucoma. Investig. Ophthalmol. Vis. Sci. 2011, 52, 4626–4629. [Google Scholar] [CrossRef]
  90. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Proteomics. Tissue-Based Map of the Human Proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  91. Thul, P.J.; Åkesson, L.; Wiking, M.; Mahdessian, D.; Geladaki, A.; Ait Blal, H.; Alm, T.; Asplund, A.; Björk, L.; Breckels, L.M.; et al. A Subcellular Map of the Human Proteome. Science 2017, 356, eaal3321. [Google Scholar] [CrossRef] [PubMed]
  92. Nakamura, J.; Meguro, A.; Ota, M.; Nomura, E.; Nishide, T.; Kashiwagi, K.; Mabuchi, F.; Iijima, H.; Kawase, K.; Yamamoto, T.; et al. Association of Toll-like Receptor 2 Gene Polymorphisms with Normal Tension Glaucoma. Mol. Vis. 2009, 15, 2905–2910. [Google Scholar] [PubMed]
  93. Shibuya, E.; Meguro, A.; Ota, M.; Kashiwagi, K.; Mabuchi, F.; Iijima, H.; Kawase, K.; Yamamoto, T.; Nakamura, M.; Negi, A.; et al. Association of Toll-like Receptor 4 Gene Polymorphisms with Normal Tension Glaucoma. Investig. Ophthalmol. Vis. Sci. 2008, 49, 4453–4457. [Google Scholar] [CrossRef] [PubMed]
  94. Takano, Y.; Shi, D.; Shimizu, A.; Funayama, T.; Mashima, Y.; Yasuda, N.; Fukuchi, T.; Abe, H.; Ideta, H.; Zheng, X.; et al. Association of Toll-like Receptor 4 Gene Polymorphisms in Japanese Subjects with Primary Open-Angle, Normal-Tension, and Exfoliation Glaucoma. Am. J. Ophthalmol. 2012, 154, 825–832.e1. [Google Scholar] [CrossRef]
  95. Bellaye, P.-S.; Shimbori, C.; Sato, S.; Upagupta, C.; Ask, K.; Gauldie, J.; Kolb, M.R.J. Loxl1 Null Mice Are Protected from TGF-β1 Induced Pulmonary Fibrosis. In D21. Immune Pathways in Acute Lung Injury and Fibrosis; American Thoracic Society International Conference Abstracts; American Thoracic Society: New York, NY, USA, 2017; p. 7079. [Google Scholar]
  96. Yang, A.; Yan, X.; Fan, X.; Shi, Y.; Huang, T.; Li, W.; Chen, W.; Jia, J.; You, H. Hepatic Stellate Cells-Specific LOXL1 Deficiency Abrogates Hepatic Inflammation, Fibrosis, and Corrects Lipid Metabolic Abnormalities in Non-Obese NASH Mice. Hepatol. Int. 2021, 15, 1122–1135. [Google Scholar] [CrossRef]
  97. Tanito, M.; Minami, M.; Akahori, M.; Kaidzu, S.; Takai, Y.; Ohira, A.; Iwata, T. LOXL1 Variants in Elderly Japanese Patients with Exfoliation Syndrome/Glaucoma, Primary Open-Angle Glaucoma, Normal Tension Glaucoma, and Cataract. Mol. Vis. 2008, 14, 1898–1905. [Google Scholar]
  98. Wolf, C.; Gramer, E.; Müller-Myhsok, B.; Pasutto, F.; Gramer, G.; Wissinger, B.; Weisschuh, N. Lysyl Oxidase-like 1 Gene Polymorphisms in German Patients with Normal Tension Glaucoma, Pigmentary Glaucoma and Exfoliation Glaucoma. J. Glaucoma 2010, 19, 136–141. [Google Scholar] [CrossRef]
  99. Lu, S.Y.; Rong, S.S.; Wu, Z.; Huang, C.; Matsushita, K.; Ng, T.K.; Leung, C.K.S.; Kawashima, R.; Usui, S.; Tam, P.O.S.; et al. Association of the CAV1-CAV2 Locus with Normal-Tension Glaucoma in Chinese and Japanese. Clin. Experiment. Ophthalmol. 2020, 48, 658–665. [Google Scholar] [CrossRef]
  100. Li, X.; Gu, X.; Boyce, T.M.; Zheng, M.; Reagan, A.M.; Qi, H.; Mandal, N.; Cohen, A.W.; Callegan, M.C.; Carr, D.J.J.; et al. Caveolin-1 Increases Proinflammatory Chemoattractants and Blood-Retinal Barrier Breakdown but Decreases Leukocyte Recruitment in Inflammation. Investig. Ophthalmol. Vis. Sci. 2014, 55, 6224–6234. [Google Scholar] [CrossRef]
  101. Reagan, A.; Gu, X.; Hauck, S.M.; Ash, J.D.; Cao, G.; Thompson, T.C.; Elliott, M.H. Retinal Caveolin-1 Modulates Neuroprotective Signaling. In Retinal Degenerative Diseases; Springer International Publishing: Berlin/Heidelberg, Germany, 2016; pp. 411–418. [Google Scholar]
  102. Carnes, M.U.; Liu, Y.P.; Allingham, R.R.; Whigham, B.T.; Havens, S.; Garrett, M.E.; Qiao, C.; NEIGHBORHOOD Consortium Investigators; Katsanis, N.; Wiggs, J.L.; et al. Discovery and Functional Annotation of SIX6 Variants in Primary Open-Angle Glaucoma. PLoS Genet. 2014, 10, e1004372. [Google Scholar]
  103. Shin, H.-Y.; Lee, Y.C.; Lee, M.Y. Association of Polymorphisms at the SIX1/SIX6 Locus With Normal Tension Glaucoma in a Korean Population. J. Glaucoma 2022, 31, 763–766. [Google Scholar] [CrossRef] [PubMed]
  104. Skowronska-Krawczyk, D.; Zhao, L.; Zhu, J.; Weinreb, R.N.; Cao, G.; Luo, J.; Flagg, K.; Patel, S.; Wen, C.; Krupa, M.; et al. P16INK4a Upregulation Mediated by SIX6 Defines Retinal Ganglion Cell Pathogenesis in Glaucoma. Mol. Cell 2015, 59, 931–940. [Google Scholar] [CrossRef] [PubMed]
  105. Bussian, T.J.; Aziz, A.; Meyer, C.F.; Swenson, B.L.; van Deursen, J.M.; Baker, D.J. Clearance of Senescent Glial Cells Prevents Tau-Dependent Pathology and Cognitive Decline. Nature 2018, 562, 578–582. [Google Scholar] [CrossRef] [PubMed]
  106. Lu, S.Y.; He, Z.Z.; Xu, J.X.; Yang, C.; Chen, L.J.; Gong, B. Association of Polymorphisms at the SIX1-SIX6 Locus With Primary Open-Angle Glaucoma. Investig. Ophthalmol. Vis. Sci. 2019, 60, 2914–2924. [Google Scholar] [CrossRef]
  107. Adrados, I.; Larrasa-Alonso, J.; Galarreta, A.; López-Antona, I.; Menéndez, C.; Abad, M.; Gil, J.; Moreno-Bueno, G.; Palmero, I. The Homeoprotein SIX1 Controls Cellular Senescence through the Regulation of P16INK4A and Differentiation-Related Genes. Oncogene 2016, 35, 3485–3494. [Google Scholar] [CrossRef] [PubMed]
  108. Liu, Z.; Mar, K.B.; Hanners, N.W.; Perelman, S.S.; Kanchwala, M.; Xing, C.; Schoggins, J.W.; Alto, N.M. A NIK-SIX Signalling Axis Controls Inflammation by Targeted Silencing of Non-Canonical NF-ΚB. Nature 2019, 568, 249–253. [Google Scholar] [CrossRef]
  109. Khakh, B.S.; McCarthy, K.D. Astrocyte Calcium Signaling: From Observations to Functions and the Challenges Therein. Cold Spring Harb. Perspect. Biol. 2015, 7, a020404. [Google Scholar] [CrossRef]
  110. Semyanov, A.; Henneberger, C.; Agarwal, A. Making Sense of Astrocytic Calcium Signals—From Acquisition to Interpretation. Nat. Rev. Neurosci. 2020, 21, 551–564. [Google Scholar] [CrossRef]
  111. Wang, Q.-C.; Zheng, Q.; Tan, H.; Zhang, B.; Li, X.; Yang, Y.; Yu, J.; Liu, Y.; Chai, H.; Wang, X.; et al. TMCO1 Is an ER Ca2+ Load-Activated Ca2+ Channel. Cell 2016, 165, 1454–1466. [Google Scholar] [CrossRef] [Green Version]
  112. Sun, Z.; Zhang, H.; Wang, X.; Wang, Q.-C.; Zhang, C.; Wang, J.-Q.; Wang, Y.-H.; An, C.-Q.; Yang, K.-Y.; Wang, Y.; et al. TMCO1 Is Essential for Ovarian Follicle Development by Regulating ER Ca2+ Store of Granulosa Cells. Cell Death Differ. 2018, 25, 1686–1701. [Google Scholar] [CrossRef]
  113. Shigetomi, E.; Saito, K.; Sano, F.; Koizumi, S. Aberrant Calcium Signals in Reactive Astrocytes: A Key Process in Neurological Disorders. Int. J. Mol. Sci. 2019, 20, 996. [Google Scholar] [CrossRef]
  114. Gharahkhani, P.; Jorgenson, E.; Hysi, P.; Khawaja, A.P.; Pendergrass, S.; Han, X.; Ong, J.S.; Hewitt, A.W.; Segrè, A.V.; Rouhana, J.M.; et al. Genome-Wide Meta-Analysis Identifies 127 Open-Angle Glaucoma Loci with Consistent Effect across Ancestries. Nat. Commun. 2021, 12, 1258. [Google Scholar] [CrossRef]
  115. Sompol, P.; Furman, J.L.; Pleiss, M.M.; Kraner, S.D.; Artiushin, I.A.; Batten, S.R.; Quintero, J.E.; Simmerman, L.A.; Beckett, T.L.; Lovell, M.A.; et al. Calcineurin/NFAT Signaling in Activated Astrocytes Drives Network Hyperexcitability in Aβ-Bearing Mice. J. Neurosci. 2017, 37, 6132–6148. [Google Scholar] [CrossRef] [PubMed]
  116. Nakase, T.; Söhl, G.; Theis, M.; Willecke, K.; Naus, C.C.G. Increased Apoptosis and Inflammation after Focal Brain Ischemia in Mice Lacking Connexin43 in Astrocytes. Am. J. Pathol. 2004, 164, 2067–2075. [Google Scholar] [CrossRef]
  117. Hernandez, M.R.; Miao, H.; Lukas, T. Astrocytes in Glaucomatous Optic Neuropathy. Prog. Brain Res. 2008, 173, 353–373. [Google Scholar]
  118. Kerr, N.M.; Johnson, C.S.; Green, C.R.; Danesh-Meyer, H.V. Gap Junction Protein Connexin43 (GJA1) in the Human Glaucomatous Optic Nerve Head and Retina. J. Clin. Neurosci. 2011, 18, 102–108. [Google Scholar] [CrossRef] [PubMed]
  119. Malone, P.; Miao, H.; Parker, A.; Juarez, S.; Hernandez, M.R. Pressure Induces Loss of Gap Junction Communication and Redistribution of Connexin 43 in Astrocytes. Glia 2007, 55, 1085–1098. [Google Scholar] [CrossRef] [PubMed]
  120. Hernandez, M.R. The Optic Nerve Head in Glaucoma: Role of Astrocytes in Tissue Remodeling. Prog. Retin. Eye Res. 2000, 19, 297–321. [Google Scholar] [CrossRef]
  121. Dai, C.; Khaw, P.T.; Yin, Z.Q.; Li, D.; Raisman, G.; Li, Y. Structural Basis of Glaucoma: The Fortified Astrocytes of the Optic Nerve Head Are the Target of Raised Intraocular Pressure. Glia 2012, 60, 13–28. [Google Scholar] [CrossRef]
  122. Morgan, J.E. Optic Nerve Head Structure in Glaucoma: Astrocytes as Mediators of Axonal Damage. Eye 2000, 14 Pt 3B, 437–444. [Google Scholar] [CrossRef] [Green Version]
  123. Pena, J.D.; Varela, H.J.; Ricard, C.S.; Hernandez, M.R. Enhanced Tenascin Expression Associated with Reactive Astrocytes in Human Optic Nerve Heads with Primary Open Angle Glaucoma. Exp. Eye Res. 1999, 68, 29–40. [Google Scholar] [CrossRef] [PubMed]
  124. Varela, H.J.; Hernandez, M.R. Astrocyte Responses in Human Optic Nerve Head with Primary Open-Angle Glaucoma. J. Glaucoma 1997, 6, 303–313. [Google Scholar] [CrossRef]
  125. Yuan, L.; Neufeld, A.H. Tumor Necrosis Factor-Alpha: A Potentially Neurodestructive Cytokine Produced by Glia in the Human Glaucomatous Optic Nerve Head. Glia 2000, 32, 42–50. [Google Scholar] [CrossRef] [PubMed]
  126. Hernandez, M.R.; Pena, J.D. The Optic Nerve Head in Glaucomatous Optic Neuropathy. Arch. Ophthalmol. 1997, 115, 389–395. [Google Scholar] [CrossRef] [PubMed]
  127. Chidlow, G.; Ebneter, A.; Wood, J.P.M.; Casson, R.J. The Optic Nerve Head Is the Site of Axonal Transport Disruption, Axonal Cytoskeleton Damage and Putative Axonal Regeneration Failure in a Rat Model of Glaucoma. Acta Neuropathol. 2011, 121, 737–751. [Google Scholar] [CrossRef] [PubMed]
  128. Elkington, A.R.; Inman, C.B.; Steart, P.V.; Weller, R.O. The Structure of the Lamina Cribrosa of the Human Eye: An Immunocytochemical and Electron Microscopical Study. Eye 1990, 4 Pt 1, 42–57. [Google Scholar] [CrossRef] [PubMed]
  129. Cooper, M.L.; Collyer, J.W.; Calkins, D.J. Astrocyte Remodeling without Gliosis Precedes Optic Nerve Axonopathy. Acta Neuropathol. Commun. 2018, 6, 38. [Google Scholar] [CrossRef]
  130. Yan, X.; Tezel, G.; Wax, M.B.; Edward, D.P. Matrix Metalloproteinases and Tumor Necrosis Factor Alpha in Glaucomatous Optic Nerve Head. Arch. Ophthalmol. 2000, 118, 666–673. [Google Scholar] [CrossRef] [PubMed]
  131. Fuchshofer, R. The Pathogenic Role of Transforming Growth Factor-Β2 in Glaucomatous Damage to the Optic Nerve Head. Exp. Eye Res. 2011, 93, 165–169. [Google Scholar] [CrossRef]
  132. Zode, G.S.; Clark, A.F.; Wordinger, R.J. Bone Morphogenetic Protein 4 Inhibits TGF-Beta2 Stimulation of Extracellular Matrix Proteins in Optic Nerve Head Cells: Role of Gremlin in ECM Modulation. Glia 2009, 57, 755–766. [Google Scholar] [CrossRef]
  133. Neumann, C.; Yu, A.; Welge-Lüssen, U.; Lütjen-Drecoll, E.; Birke, M. The Effect of TGF-Beta2 on Elastin, Type VI Collagen, and Components of the Proteolytic Degradation System in Human Optic Nerve Astrocytes. Investig. Ophthalmol. Vis. Sci. 2008, 49, 1464–1472. [Google Scholar] [CrossRef] [PubMed]
  134. Kim, M.-L.; Sung, K.R.; Kwon, J.; Shin, J.A. Statins Suppress TGF-Β2-Mediated MMP-2 and MMP-9 Expression and Activation Through RhoA/ROCK Inhibition in Astrocytes of the Human Optic Nerve Head. Investig. Ophthalmol. Vis. Sci. 2020, 61, 29. [Google Scholar] [CrossRef] [PubMed]
  135. Oram, J.F.; Heinecke, J.W. ATP-Binding Cassette Transporter A1: A Cell Cholesterol Exporter That Protects against Cardiovascular Disease. Physiol. Rev. 2005, 85, 1343–1372. [Google Scholar] [CrossRef]
  136. Yu, A.L.; Welge-Lussen, U. Antioxidants Reduce TGF-Beta2-Induced Gene Expressions in Human Optic Nerve Head Astrocytes. Acta Ophthalmol. 2013, 91, e92–e98. [Google Scholar] [CrossRef] [PubMed]
  137. Shinozaki, Y.; Koizumi, S.; Ohno, Y.; Nagao, T.; Inoue, K. Extracellular ATP Counteracts the ERK1/2-Mediated Death-Promoting Signaling Cascades in Astrocytes. Glia 2006, 54, 606–618. [Google Scholar] [CrossRef]
  138. Shinozaki, Y.; Koizumi, S.; Ishida, S.; Sawada, J.-I.; Ohno, Y.; Inoue, K. Cytoprotection against Oxidative Stress-Induced Damage of Astrocytes by Extracellular ATP via P2Y1 Receptors. Glia 2005, 49, 288–300. [Google Scholar] [CrossRef]
  139. Hamada, K.; Shinozaki, Y.; Namekata, K. Loss of P2Y1 Receptors Triggers Glaucoma-like Pathology in Mice. Br. J. Pharmacol. 2021, 178, 4552–4571. [Google Scholar] [CrossRef]
  140. Neufeld, A.H.; Hernandez, M.R.; Gonzalez, M. Nitric Oxide Synthase in the Human Glaucomatous Optic Nerve Head. Arch. Ophthalmol. 1997, 115, 497–503. [Google Scholar] [CrossRef] [PubMed]
  141. Liu, B.; Neufeld, A.H. Expression of Nitric Oxide Synthase-2 (NOS-2) in Reactive Astrocytes of the Human Glaucomatous Optic Nerve Head. Glia 2000, 30, 178–186. [Google Scholar] [CrossRef]
  142. Neufeld, A.H. Nitric Oxide: A Potential Mediator of Retinal Ganglion Cell Damage in Glaucoma. Surv. Ophthalmol. 1999, 43 (Suppl. 1), S129–S135. [Google Scholar] [CrossRef] [PubMed]
  143. Morgan, J.; Caprioli, J.; Koseki, Y. Nitric Oxide Mediates Excitotoxic and Anoxic Damage in Rat Retinal Ganglion Cells Cocultured with Astroglia. Arch. Ophthalmol. 1999, 117, 1524–1529. [Google Scholar] [CrossRef]
  144. Fortune, B.; Ma, K.N.; Gardiner, S.K.; Demirel, S.; Mansberger, S.L. Peripapillary Retinoschisis in Glaucoma: Association with Progression and OCT Signs of Müller Cell Involvement. Investig. Ophthalmol. Vis. Sci. 2018, 59, 2818–2827. [Google Scholar] [CrossRef] [PubMed]
  145. Tezel, G.; Li, L.Y.; Patil, R.V.; Wax, M.B. TNF-α and TNF-α Receptor-1 in the Retina of Normal and Glaucomatous Eyes. Investig. Ophthalmol. Vis. Sci. 2001, 42, 1787–1794. [Google Scholar]
  146. Schmidt, A.M.; Yan, S.D.; Wautier, J.L.; Stern, D. Activation of Receptor for Advanced Glycation End Products: A Mechanism for Chronic Vascular Dysfunction in Diabetic Vasculopathy and Atherosclerosis. Circ. Res. 1999, 84, 489–497. [Google Scholar] [CrossRef]
  147. Tezel, G.; Luo, C.; Yang, X. Accelerated Aging in Glaucoma: Immunohistochemical Assessment of Advanced Glycation End Products in the Human Retina and Optic Nerve Head. Investig. Ophthalmol. Vis. Sci. 2007, 48, 1201–1211. [Google Scholar] [CrossRef] [PubMed]
  148. Fin, N.A.S.; Sukkar, M.; Golzan, M. Receptor for Advanced Glycation End Product (RAGE) Mediates Retinal Ganglion Cell Loss in Experimental Glaucoma. Investig. Ophthalmol. Vis. Sci. 2018, 59, 3726. [Google Scholar]
  149. Grieshaber, M.C.; Orgul, S.; Schoetzau, A.; Flammer, J. Relationship between Retinal Glial Cell Activation in Glaucoma and Vascular Dysregulation. J. Glaucoma 2007, 16, 215–219. [Google Scholar] [CrossRef]
  150. Grieshaber, M.C.; Moramarco, F.; Schoetzau, A.; Flammer, J.; Orguel, S. Detection of Retinal Glial Cell Activation in Glaucoma by Time Domain Optical Coherence Tomography. Klin. Monbl. Augenheilkd. 2012, 229, 314–318. [Google Scholar] [CrossRef]
  151. Sugiyama, T.; Moriya, S.; Oku, H.; Azuma, I. Association of Endothelin-1 with Normal Tension Glaucoma: Clinical and Fundamental Studies. Surv. Ophthalmol. 1995, 39 (Suppl. S1), S49–S56. [Google Scholar] [CrossRef]
  152. Kaiser, H.J.; Flammer, J.; Wenk, M.; Lüscher, T. Endothelin-1 Plasma Levels in Normal-Tension Glaucoma: Abnormal Response to Postural Changes. Graefes Arch. Clin. Exp. Ophthalmol. 1995, 233, 484–488. [Google Scholar] [CrossRef] [PubMed]
  153. Kim, S.H.; Kim, J.Y.; Kim, D.M.; Ko, H.S.; Kim, S.Y.; Yoo, T.; Hwang, S.S.; Park, S.S. Investigations on the Association between Normal Tension Glaucoma and Single Nucleotide Polymorphisms of the Endothelin-1 and Endothelin Receptor Genes. Mol. Vis. 2006, 12, 1016–1021. [Google Scholar] [PubMed]
  154. Wróbel-Dudzińska, D.; Kosior-Jarecka, E.; Łukasik, U.; Kocki, J.; Witczak, A.; Mosiewicz, J.; Żarnowski, T. Risk Factors in Normal-Tension Glaucoma and High-Tension Glaucoma in Relation to Polymorphisms of Endothelin-1 Gene and Endothelin-1 Receptor Type A Gene. J. Ophthalmol. 2015, 2015, 368792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Ishikawa, K.; Funayama, T.; Ohtake, Y.; Kimura, I.; Ideta, H.; Nakamoto, K.; Yasuda, N.; Fukuchi, T.; Fujimaki, T.; Murakami, A.; et al. Association between Glaucoma and Gene Polymorphism of Endothelin Type A Receptor. Mol. Vis. 2005, 11, 431–437. [Google Scholar]
  156. Kosior-Jarecka, E.; Wróbel-Dudzińska, D.; Łukasik, U.; Aung, T.; Khor, C.C.; Kocki, J.; Żarnowski, T. Plasma Endothelin-1 and Single Nucleotide Polymorphisms of Endothelin-1 and Endothelin Type A Receptor Genes as Risk Factors for Normal Tension Glaucoma. Mol. Vis. 2016, 22, 1256–1266. [Google Scholar]
  157. Wu, X.; Konieczka, K.; Liu, X.; Chen, M.; Yao, K.; Wang, K.; Flammer, J. Role of Ocular Blood Flow in Normal Tension Glaucoma. Adv. Ophthalmol. Pract. Res. 2022, 2, 100036. [Google Scholar] [CrossRef]
  158. Furlanetto, R.L.; De Moraes, C.G.; Teng, C.C.; Liebmann, J.M.; Greenfield, D.S.; Gardiner, S.K.; Ritch, R.; Krupin, T. Low-Pressure Glaucoma Treatment Study Group Risk Factors for Optic Disc Hemorrhage in the Low-Pressure Glaucoma Treatment Study. Am. J. Ophthalmol. 2014, 157, 945–952. [Google Scholar] [CrossRef]
  159. Shields, M.B. Normal-Tension Glaucoma: Is It Different from Primary Open-Angle Glaucoma? Curr. Opin. Ophthalmol. 2008, 19, 85–88. [Google Scholar] [CrossRef]
  160. Prasanna, G.; Krishnamoorthy, R.; Clark, A.F.; Wordinger, R.J.; Yorio, T. Human Optic Nerve Head Astrocytes as a Target for Endothelin-1. Investig. Ophthalmol. Vis. Sci. 2002, 43, 2704–2713. [Google Scholar]
  161. Alrashdi, S.F.; Deliyanti, D.; Talia, D.M.; Wilkinson-Berka, J.L. Endothelin-2 Injures the Blood-Retinal Barrier and Macroglial Müller Cells: Interactions with Angiotensin II, Aldosterone, and NADPH Oxidase. Am. J. Pathol. 2018, 188, 805–817. [Google Scholar] [CrossRef]
  162. Williams, P.A.; Tribble, J.R.; Pepper, K.W.; Cross, S.D.; Morgan, B.P.; Morgan, J.E.; John, S.W.M.; Howell, G.R. Inhibition of the Classical Pathway of the Complement Cascade Prevents Early Dendritic and Synaptic Degeneration in Glaucoma. Mol. Neurodegener. 2016, 11, 26. [Google Scholar] [CrossRef]
  163. Howell, G.R.; Macalinao, D.G.; Sousa, G.L.; Walden, M.; Soto, I.; Kneeland, S.C.; Barbay, J.M.; King, B.L.; Marchant, J.K.; Hibbs, M.; et al. Molecular Clustering Identifies Complement and Endothelin Induction as Early Events in a Mouse Model of Glaucoma. J. Clin. Investig. 2011, 121, 1429–1444. [Google Scholar] [CrossRef] [PubMed]
  164. He, S.; Prasanna, G.; Yorio, T. Endothelin-1-Mediated Signaling in the Expression of Matrix Metalloproteinases and Tissue Inhibitors of Metalloproteinases in Astrocytes. Investig. Ophthalmol. Vis. Sci. 2007, 48, 3737–3745. [Google Scholar] [CrossRef] [PubMed]
  165. McGwin, G., Jr.; McNeal, S.; Owsley, C.; Girkin, C.; Epstein, D.; Lee, P.P. Statins and Other Cholesterol-Lowering Medications and the Presence of Glaucoma. Arch. Ophthalmol. 2004, 122, 822–826. [Google Scholar] [CrossRef] [PubMed]
  166. Boal, A.M.; Risner, M.L.; Cooper, M.L.; Wareham, L.K.; Calkins, D.J. Astrocyte Networks as Therapeutic Targets in Glaucomatous Neurodegeneration. Cells 2021, 10, 1368. [Google Scholar] [CrossRef] [PubMed]
  167. Li, S.; Jakobs, T.C. Secreted Phosphoprotein 1 Slows Neurodegeneration and Rescues Visual Function in Mouse Models of Aging and Glaucoma. Cell Rep. 2022, 41, 111880. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Elevated intraocular pressure (IOP) and glaucoma.
Figure 1. Elevated intraocular pressure (IOP) and glaucoma.
Ijms 24 02747 g001
Figure 2. Cupping of the optic nerve head.
Figure 2. Cupping of the optic nerve head.
Ijms 24 02747 g002
Figure 3. Glial cells in the retina.
Figure 3. Glial cells in the retina.
Ijms 24 02747 g003
Figure 4. Astrocyte-lineage cells are closely associated with retinal ganglion cell (RGC) axons. (a) Retinal astrocytes (magenta) attach to Tuj1-positive RGC axons (green). (b) YC-Nano-expressing Müller cells [36] (magenta) elongate their processes toward and surround RGC soma (green, asterisk). A part of the Müller cell processes enwraps SMI32-positive RGC axons (arrow). (c) Vertical section of the optical nerve head (ONH). GFAP-positive astrocytes are highly enriched and closely attached to the optic nerve. (d) Horizontal section of the ONH: (left) an astrocyte forms a honeycomb structure and (right) RGC axons pass through the hole.
Figure 4. Astrocyte-lineage cells are closely associated with retinal ganglion cell (RGC) axons. (a) Retinal astrocytes (magenta) attach to Tuj1-positive RGC axons (green). (b) YC-Nano-expressing Müller cells [36] (magenta) elongate their processes toward and surround RGC soma (green, asterisk). A part of the Müller cell processes enwraps SMI32-positive RGC axons (arrow). (c) Vertical section of the optical nerve head (ONH). GFAP-positive astrocytes are highly enriched and closely attached to the optic nerve. (d) Horizontal section of the ONH: (left) an astrocyte forms a honeycomb structure and (right) RGC axons pass through the hole.
Ijms 24 02747 g004
Figure 5. Glaucoma-related immune responses by astrocyte-lineage cells.
Figure 5. Glaucoma-related immune responses by astrocyte-lineage cells.
Ijms 24 02747 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shinozaki, Y.; Kashiwagi, K.; Koizumi, S. Astrocyte Immune Functions and Glaucoma. Int. J. Mol. Sci. 2023, 24, 2747. https://doi.org/10.3390/ijms24032747

AMA Style

Shinozaki Y, Kashiwagi K, Koizumi S. Astrocyte Immune Functions and Glaucoma. International Journal of Molecular Sciences. 2023; 24(3):2747. https://doi.org/10.3390/ijms24032747

Chicago/Turabian Style

Shinozaki, Youichi, Kenji Kashiwagi, and Schuichi Koizumi. 2023. "Astrocyte Immune Functions and Glaucoma" International Journal of Molecular Sciences 24, no. 3: 2747. https://doi.org/10.3390/ijms24032747

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop