You are on page 1of 17

Reviews

The Pathogenesis of Glaucoma in the Interplay with the


Immune System
Jochen Rieck

Glaucoma was previously thought to be caused only through is understood only in part; as a result, useful mass screening for
an elevated intraocular pressure as a sole trigger. Emerging glaucoma is lacking with respect to reliable yet affordable and
evidence indicates that the pathogenesis of glaucoma depends expeditious diagnosis.24
on several interacting pathogenetic mechanisms, which Undisputedly, a permanent raised individual ocular pressure
include mechanical effects by an increased IOP, decreased above 21 mm Hg is an important pathogenic factor in the
neutrophine-supply, hypoxia, excitotoxicity, oxidative stress disease risk.9,10,25–27 Individual ocular pressure constitutes a
and the involvement of autoimmune processes. These autoim- person-specific variation and follows a circadian rhythm.28
mune processes within the central nervous system are a highly Following this rhythm, the IOP oscillates between 10 and 21
organized response of the innate immunity. Through the mm Hg in healthy adults. Newborns have an IOP between 6
recognition of neuronal epitopes, the long-term induction of and 8 mm Hg, which rises approximately 1 mm Hg every 2
the innate immune response and its transition to an adaptive years.25,26,29–34
form might be central to the pathophysiology of the glaucoma In addition, there are healthy people who have an IOP
disease. Regardless of the pathogenic mechanism, the conse- significantly elevated above normal without ever developing
quences are always the establishment of extensive degenera- glaucoma. This condition is known as ocular hypertension
tive processes in the optic nerve head, the retinal ganglion (OHT). In direct contrast, many patients with an IOP  21 mm
cells and the axons of the optic nerve, which will lead in the Hg progress to glaucoma. Accordingly, this form of glaucoma is
irreversible destruction of these neurons. This review article called normal-tension glaucoma (NTG). Important for the
summarizes the current knowledge concerning the pathogen- pathogenesis of glaucoma therefore seems to be the individual,
esis of glaucoma, with special focus on its interplay with the relative value of IOP.
immune system. (Invest Ophthalmol Vis Sci. 2013;54:2393–
2409) DOI:10.1167/iovs.12-9781
AVAILABLE TREATMENT OPTIONS FOR GLAUCOMA
G laucoma, behind cataracts, is the second leading cause of
blindness worldwide. For the year 2020 it is expected that
approximately 80 million people will suffer from glaucoma,
The treatment of glaucoma is almost always aimed at reducing
the IOP, which leads, surprisingly, even in patients with NTG, to
which is anticipated to result in 11.2 million cases of bilateral an alleviation of the disease process.4,13,15,35–39 The reduction
blindness.1 of IOP is obtained by eye drops or systemic application of
Nowadays glaucoma is a collective term for a group of glaucoma medications. These include carbonic anhydrase
neurodegenerative processes affecting the entire visual path- inhibitors, beta-blockers, cholinergic agonists, a2-adrenoceptor
way, characterized by progressive, irreversible destruction and agonists and prostaglandins.25,27,37,40–43 Laser therapy or
death of retinal ganglion cells.2–15 These neurodegenerative surgical procedures for ensuring an adequate aqueous humor
processes are associated with a progressive visual field loss, outflow are seen as a last resort to use.44
which typically begins with an arcuate Bjerrum scotoma in the
central visual field and ends with the total blindness of the
eye.13,16–23 PATHOGENESIS AND PROGRESSION OF GLAUCOMA
Well-marked symptoms are known only in acute angle-
closure glaucoma; all other forms of chronic glaucoma are For decades, a permanent raised IOP over 21 mm Hg was
largely asymptomatic. This is the main reason the disease considered the sole trigger for the onset of glaucoma. In fact, in
accomplishes its destruction to a far extent unnoticed. As many the most common glaucoma type, primary open-angle
as 50% of all affected patients live without diagnosis until glaucoma (POAG), the majority of those affected exhibit a
advanced illness.13 A simplified diagnostic procedure, ideally a raised IOP. Therefore this glaucoma subtype belongs to the
rapid glaucoma test that could be carried out by an optician, group of high-tension glaucomas (HTG). However, approxi-
would be an effective tool to gain time through early diagnosis mately one-third of all POAG patients had at no time a
and treatment. However, to date the pathogenesis of glaucoma pathologically elevated IOP (NTG patients); this raises the
question what, if not the IOP, is responsible for the destruction
of retinal ganglion cells.25,27,45–47 It is worth mentioning that
therapeutic lowering of the IOP leads to a slowing of disease
From the Institut für Zoologie, Johannes Gutenberg-Universität progression in a subgroup of NTG patients.43,48–50
Mainz, Mainz, Germany.
The substantial demise of retinal ganglion cells (RGCs) is
Submitted for publication March 1, 2012; revised August 19 and
December 17, 2012; accepted January 26, 2013. accompanied by morphologic changes of the retina, of which
Disclosure: J. Rieck, None the cupping of the optic nerve head (ONH) is the most
Corresponding author: Jochen Rieck, Institut für Zoologie, prominent. Thereby glaucomatous damage is not confined just
Johannes Gutenberg-Universität Mainz, Johannes von Müller-Weg 6, to the RGCs and their axons, somata, and dendrites. Rather the
55099 Mainz, Germany; rieck@uni-mainz.de. entire visual pathway from the retina to the visual cortex in the

DOI:10.1167/iovs.12-9781
Investigative Ophthalmology & Visual Science, March 2013, Vol. 54, No. 3
Copyright 2013 The Association for Research in Vision and Ophthalmology, Inc. 2393

Downloaded from iovs.arvojournals.org on 02/05/2019


2394 Rieck IOVS, March 2013, Vol. 54, No. 3

brain is affected.14,26,51–54 However, it is not clear whether the THE DEMISE OF RETINAL GANGLION CELLS AS A
remodeling of the ONH leads to the destruction of RGCs, or
conversely, whether the RGC loss in glaucoma is causative for CENTRAL HALLMARK OF GLAUCOMA PATHOGENESIS
the characteristic cupping of the ONH.8,9,15,30,50,55,56 Beyond In glaucoma, it seems that the nature and position of the
controversy, apoptosis is accepted as an important component injurious trigger determine which path is taken into neurode-
of glaucomatous neurodegeneration.6,10,12,13,17,19,20,31,36,50,57–64 generation. Already documented is the initiation of pro-
The involvement of apoptotic processes, as well as the grammed cell death of RGCs via tumor suppressor protein
correlation between IOP and glaucomatous damage, is rather p53, but also through the direct activation of the ‘‘death
obvious in pathologic conditions with rapidly raised IOP, as it receptor’’ CD95 in autoreactive conditions.58,63,93–95
can be monitored in acute angle-closure glaucomas and acute In general, the RGC’s fate depends on multiple trophic as
secondary glaucomas.12,15,20,65–67 As demonstrated in animal well as degenerative signaling pathways within the soma and/
models, severe pressure insults may cause axonal degeneration or signaling from the environment, which has the potential to
and apoptosis within hours.15,31,33,68 disrupt neuronal homeostasis. One example is the cytotoxic
However, in open-angle glaucomas and mild forms of effect of b-amyloid protein, which causes apoptosis by binding
secondary glaucomas, the correlation between IOP and to neurotrophin receptor p75NTR or through the accumulation
neuronal degeneration is not so apparent. OHT patients with of protein aggregates, as in Alzheimer’s disease.96,97 p75NTR,
slightly elevated IOP between 21 and 30 mm Hg, without another known cause, is involved in light-induced photore-
detectable signs of disc cupping and/or visual field defects, do ceptor apoptosis.98,99
not necessarily progress to glaucoma.69 The Ocular Hyperten- Furthermore, neurotransmitters such as dopamine, seroto-
sion Treatment Study demonstrated that over 90% of the nin, and glutamate have the potential to drive RGCs into
untreated control group did not develop any form of glaucoma programmed cell death. As a cause for the triggering of
within the 5-year study period.25,27,70 apoptosis, excitatory mechanisms are suggested.36,43,50,100–110
Regarding the potential excitotoxicity of glutamate, it is
noteworthy that glutamate transporters like GLAST and EEAC1,
THE LAMINA CRIBROSA AS THE MOST VULNERABLE as well as glutamate receptors, are downregulated in glau-
PART OF THE RETINAL GANGLION CELL comatous eyes following astrocyte activation.102,111,112 As a
consequence, deficient glutamate removal from the extracel-
As mentioned above, an elevated IOP is one of the main risk lular space might represent an interaction by which astrocyte
factors for glaucoma, which may be associated with a or glial activation can result in RGC death.102,112
deficiency in the cellular nutritional state of the RGCs. This Considering the complexity of RGCs, Whitmore et al.
presumption is suggested by an animal model in which an postulate the model of ‘‘compartmentalized self-destruction’’
elevated IOP causes the failure of the neurotrophin supply due in neurons, suggesting that depending on the nature and
to the collapse of axonal transport.71,72 location of the initial insult, somal death can be preceded by a
RGCs respond to a variety of neurotrophins, but mainly to number of degenerative processes infesting the axons,
brain-derived neurotrophic factor (BDNF), ciliary nerve trophic dentrites, and synapses of the neuron.15 These degenerative
factor (CNTF), glial cell–derived neurotrophic factor (GDNF), processes are phylogenetically highly conserved and known to
and nerve growth factor.73–77 In animal models, as well as in cell be involved in a whole range of destructive processes within
culture, the neuroprotective effects of these trophic factors are the vertebral central nervous system (CNS), as well as in axons
documented.78,79 Like other neurotrophins, nerve growth of Drosophila.10,79,113–125 Since such widespread processes of
factor (NGF) and BDNF are taken up into the target cell through neuronal degeneration may affect the entire CNS, one can
receptor-mediated endocytosis. Binding partners are the high- assume that insights into neurons outside the eye are also
affinity receptors TrkA (for NGF) and TrkB (for BDNF), and a low- applicable on RGCs. Current findings support the assumption
affinity receptor, p75NTR, to which all other neurotrophins can that self-destruct processes, especially axonal degeneration as
bind. The internalization of the receptor–ligand complex seen in non-RGC neurons, also take place during glauco-
followed by the retrograde transport to the soma of the neuron ma.10,124,126 One of these self-destruct processes, which is
is coordinated by the G-proteins Rabenosyn 1 and 5 (Rab1 and documented across species boundaries, is known as Wallerian
Rab5).80–85 Immunohistochemical studies demonstrate that degeneration (WD), a common phenomenon in cases of severe
impairing neurotrophin transport from the visual thalamus to axonal damage such as transection or experimental nerve
the soma of RCGs results in their destruction by apopto- crush (i.e., nerve crush model). WD represents valid evidence
sis.20,65,72 Similar effects are observed in the optic nerve crush that axonal degeneration is a soma-independently organized
model.19,86 process that can be studied in mice carrying the Wallerian
Here the lamina cribrosa sclerae, through which the optic Degeneration Slow (WldS) gene.15,61,127–131
nerve and the central retinal artery and vein enter the bulbus This mutation results in a resistance to axonal degeneration
oculi, appears to play a key role, since it is the weakest portion while having no direct effect on somal apoptosis. During WD,
of the sclera. A raised IOP may cause an excavation at the the axonal section, which is detached from the soma by the
position where the optic nerve (papilla nervi optici) enters the injury, degenerates in a characteristic and autonomous manner,
retina.55 By dyeing RGC axons of freshly explanted eyes, beginning at the point of transection and extending almost
Morgan et al. were able to document the path of individual synchronously throughout the axon within days.124,132,133
axons through the lamina cribrosa: Most axons take a direct A second frequently observed pathomechanism, so-called
path toward the ONH, while others, approximately 8% to 12%, dying back (DB), seems to be a variation of WD and follows
squeeze through collagenous plates. An increased IOP leads generalized neuronal stress without acute neuronal trauma.
directly to a contusion of these nerve fibers.87 Experiments with Differing from WD, DB is initiated by a withdrawal of the
primates show that the retrograde and anterograde neuronal synapses, followed by an asynchronous progression of the
transport in glaucomatous eyes is disturbed or even interrupted destruction process toward the cell soma.
at just this point.65,71,72,88–91 Additionally, the cerebrospinal In cases of DB, the neuron seems to have a distinct chance
fluid pressure influences the degree of mechanical stress, due to to regenerate if the neuronal stress decreases, whereas in WD,
a pressure gradient across the tissue of the ONH.92 somal apoptosis appears to be unavoidable.15,134,135 The

Downloaded from iovs.arvojournals.org on 02/05/2019


IOVS, March 2013, Vol. 54, No. 3 A Summary of the Current Knowledge 2395

review article by Whitmore et al.15 provides detailed informa- patients contain antibodies against neuron-specific c-enolase, a
tion concerning compartmentalized degeneration. key enzyme for glycolysis. In the healthy control group, only
The following sections provide an overview of how, on the 10% of those examined showed these antibodies. However, in
one hand, the immune system eliminates pathogens and cell glaucoma patients with increased c-enolase antibody titers,
debris to maintain the homeostasis of the CNS and, on the none of the elevated autoantibody titers against rhodopsin, a-
other hand, observations of well-studied diseases such as crystallin, and HSP60 otherwise commonly detected in
Alzheimer’s or multiple sclerosis (MS). The latter show what glaucoma patients could be demonstrated.8,146,153,159,160 Fur-
terrible damage the immune system is able to provoke in ther studies, published by Grus et al. and Joachim et al., also
situations of cellular stress and unphysiological conditions. revealed significant differences in the antibody profiles of
Many of the well-known neurodegenerative incidents that are glaucoma patients.161–163
caused by misdirected immune processes within the CNS (e.g., Furthermore, a significantly increased antibody reactivity
reactive astrogliosis) are also detectable in glaucoma.136 This against ONH glycosaminoglycans (GAGs), glutathione S-trans-
illustrates how homeostasis and survival of the RGC depend on ferase, alpha-fodrin, and neurofilament protein has been found
a well-balanced function of the immune system. in the sera of patients with glaucoma.163–166 In the case of anti-
GAG antibodies, a direct physiological impact is postulated due
to the change of organization and physical characteristics of
AUTOIMMUNE MECHANISMS IN THE ONSET OF GAGs located in the ONH. This may increase the susceptibility
GLAUCOMA of the ONH and the lamina cribrosa to tissue damage and
subsequently additional insult to the remaining axons.164
The first hint for an involvement of the immune system in However, in most studies, antibody titers in NTG patients are
glaucoma was delivered by Wax in 1998 with the detection of elevated, although some patients with elevated IOP show
antibodies against endogenous antigens such as heat shock decreased autoantibody reactivity as well.161
protein 60 (HSP60) in the serum of NTG patients.137 Heat An explanation for these measurable deviations in the
shock proteins (HSPs) are components of cellular defense antibody repertoire of patients with glaucoma could be found
mechanisms and are upregulated under pathophysiological in our own evolutionary history: Some proteins, such as
conditions.138 According to their molecular weight, HSPs are histones or HSP, are strongly enough conserved phylogeneti-
divided into two main families: the major HSPs (HSP90, HSP70, cally that molecular similarities between host and pathogenic
and HSP60) and the small HSPs, including HSP27. HSP60 and germs occasionally result in cross-reactivity.137,167–169
HSP90 are constitutively expressed in the CNS, whereas HSP27 Additionally or independently thereof, epitope spreading
and HSP70 are highly inducible in the retinal glia, the Müller could be another source of autoantibodies in glaucoma:
cells, and neurons by stressors such as ischemia and Typically, a humoral response begins with the attack on the
hyperthermia.139–142 However, HSP27 expression in the intact, epitope that elicited the initial immune response, spreading to
undamaged retina has been found to be limited.143 In states of other epitopes on the same antigen (intramolecular epitope
cellular stress, HSP27 and HSP70 act in an antiapoptotic spread) or to similar epitopes on other antigens (intermolec-
manner, whereas HSP60 promotes the induction of apopto- ular epitope spread). Autoimmunity may arise from epitope
sis.144,145 Direct evidence for the involvement of HSP27 in spreading on inflammation-induced posttranslational modified
glaucomatous processes comes from the detection of this self-antigens or from self-antigens that result from immune-
protein in RGC axons and ONH of affected human donor eyes, mediated apoptosis.170–172
considering that the axons and ONH are mainly battered under In many cases, the autoantigen that triggered the initial
glaucoma.146 immune response has been identified and in fact shares
In an animal model, HSP27 develops its neuroprotective sequence homology with a disease-causing antigenic pathogen
effects after axotomy and ischemia, possibly through the component. In this context, the application of epitope
inhibition of caspase-3.147–149 Furthermore, HSP70 also shows mapping has allowed detailed characterization of those
neuroprotective effects after ischemic conditions.141,150 epitopes whose autoantibodies bind in diseases such as
Since it has been documented that antibodies are able to systemic lupus erythematosus (SLE) (antispliceosome antibod-
enter the cytoplasm of intact cells, the effect of autoreactive ies), Devic’s syndrome (anti-aquaporin-4 antibodies), and NTG
immunoglobulins, in particular on cellular processes, is taken (antirhodopsin antibodies).173–175 Intriguingly, antirhodopsin
into account.151,152 Rats immunized with HSP27 show an antibodies found in sera of NTG patients also bind to viral and
elevated rate of apoptosis in the RGC, where the loss is focused bacterial epitopes.174
near the area centralis. NTG patients display the worst damage Regarding these facts, we have to keep in mind that
in the same retinal area.153,154 autoreactive antibodies can be not only destructive but also
In the case of HSP27, either a-HSP27 autoantibodies could protective. Recent findings support the hypothesis that these
protect HSP27 from premature degradation by matrix metal- antibodies contribute to the clearance of cellular damage and
loproteinases (MMPs), considering that neuroprotective HSP27 promote repair. So, the humoral immune system can be seen as
is among the MMP substrates, or this antibody may inhibit the a double-edged sword, with the potential both to destroy and
neuroprotective activity of HSP27 and thus lead directly to to heal. A repertoire of low-affine, so called natural autoanti-
increased apoptosis rates of RGC in glaucoma.142,155,156 bodies is present in every healthy human and seems to be of
Significantly increased levels of MMPs in glaucomatous retinae great significance during tissue injury when these autoanti-
support a hypothetical protective role of a-HSP27 autoanti- bodies contribute to the removal of cellular debris and support
bodies.157 tissue healing.176,177 Furthermore, humoral autoimmunity
On the other hand, the accumulation of MMPs correlates seems to have a protective role. Under discussion, among
with the release of TNF-a particularly in patients with NTG, in other functions, are the masking of self-recognition against
which MMPs are involved in the processing of the membrane- pathogenic autoantibodies, the neutralization of inflammatory
bound TNF-a precursor.158 cytokines like TNF-a through anticytokine antibodies, and an
But it’s not just about antibodies against HSPs: Documented immune-regulatory function.178–181 A decreased reactivity of
peculiarities in the antibody repertoire of glaucoma patients naturally occurring and perhaps protective autoantibodies may
have been reported by Maruyama et al. These authors therefore lead to a loss of immune protection and consequently
illustrated that the sera of approximately 20% of all glaucoma an increased risk of developing glaucoma.182

Downloaded from iovs.arvojournals.org on 02/05/2019


2396 Rieck IOVS, March 2013, Vol. 54, No. 3

MICROGLIA COORDINATE THE IMMUNE SYSTEM OF THE TNF-a, the activation of caspases has been shown, whereupon
TNF-a increases apoptosis of neurons via the inhibition of
CENTRAL NERVOUS SYSTEM insulin-like growth factor-1 (IGF-1).204,205 In addition to the
Today, three retinal glial cell types are known: The microglia, secretion of chemokines, activated microglial cells set free
subdivided into star-shaped astrocytes and specialized retinal toxins such as nitric oxide (NO) and prostaglandins, which in
Müller cells, face the macroglia. In the retina, astrocytes are turn initiate neurodegenerative processes.203,206 As profession-
localized in the ganglion cell layer, whereas Müller cells span al antigen-presenting cells, activated microglia control the
the entire retina. Microglia, ovoid or amoeboid shaped, can be transition from innate immunity to an adaptive immune
found in the ganglion cell and inner plexiform layer. The response in the CNS.206–208
macroglia provide the supply of the ganglion cells with However, in glaucoma, TNF-a is of special interest: TNF-a
neurotrophins, regulate extracellular ion concentration, and and its receptors TNFR1a and TNFR1b have been shown to be
support neuronal metabolism.183–186 In contrast, microglia act upregulated in the retinae of glaucoma patients, in whom
as phagocytes that eliminate apoptotic neurons, cell debris, increased expression of TNF receptors is predominant in the
and pathogens.187 Microglia recognize these via Toll-like ONH.158,209,210 In severely damaged ONHs, the axons of the
receptors (TLR) and react by releasing proinflammatory RGCs express TNFR1 and may be the direct target for TNF-a–
cytokines such as TNF-a and nitric oxide synthase-2 (NOS-2). mediated neurodegeneration.210 Thereby the increased TNF-a
Apoptotic cells release anti-inflammatory mediators such as expression seems to be specifically controlled. TrkC.T1, a
TGF-b prior to phagocytosis by microglia.188 Microglia truncated neurotrophin receptor isoform lacking the kinase
represent the first line of immune defense and are therefore domain, has been shown to be rapidly upregulated in
equipped with a finely structured arsenal of defense mecha- glaucomatous retinae. On closer examination, TrkC.T1 seems
nisms and a high degree of flexibility to act at the appropriate to be part of a fatal loop: Elevated IOP upregulates glial
place. They destroy microbes by releasing cytotoxic substanc- TrkC.T1 expression in glia; TrkC.T1 controls glial TNF-a
es, remove cell debris by phagocytosis, and act as antigen- production; and TNF-a causes RGC death.211
presenting cells.189 Another interesting aspect in this context is the occurrence
Apart from the normal motility behavior under physiolog- of single-nucleotide polymorphisms of the TNF-a gene, found
ical conditions, in the event of activation the cells migrate to be significantly higher in high-tension glaucoma patients.212
collectively to the site of injury. Chipped neurons usually set Ambiguous so far are the effects of those polymorphisms on
free nucleotides such as adenosine triphosphate (ATP), which the pathogenesis of glaucoma. While studies from Pakistan and
may activate microglia.190,191 After a few days, this event is Iran have shown a strong connection of POAG and pseudoex-
followed by glial proliferation that can end up in a reactive foliative glaucoma with TNF-a polymorphisms, others could
gliosis.192 The involvement of a gliosis in neurodegenerative not confirm this correlation.213–216
processes has been demonstrated in an animal model for So far, there are a number of findings of elevated cytokine
Alzheimer’s disease as well as in glaucoma.68,105,136,193–195 titers and activated glial cells within the CNS, but it is not clear
An indication of the positive chemotactic motility of what is causative and what is classified as an epiphenomenon
microglia is provided by experiments with CXCR3 knockout for the manifestation of an autoimmune disease. What seems to
mice. The chemokine CXCL10 expressed under degenerative be clear is that cellular stressors such as oxidative stress,
conditions by neurons cannot bind the microglial CXCR3 mitochondrial dysfunction, or excitotoxicity by glutamate are
receptor. This results in an accumulation of damaged and dead not sufficient as singular stressors to trigger the demise of
cells that would have been gone after a few days under normal neurons. Animal models for ALS, Alzheimer’s disease, Parkin-
son’s, and Huntington prove this assumption.203,217,218
circumstances.196 In an animal model of amyotrophic lateral
Similarly, the presence of antibodies directed against
sclerosis (ALS) and Parkinson’s disease, another chemokine,
neuronal antigens affects the cellular function of the epitope,
CX3CL1, has been identified that is released from damaged
but these are not able to trigger a neurodegenerative
neurons as a chemoattractant for glial cells. CX3CR1/ mice
autoimmune disease. Tezel et al. succeeded in inducing
whose microglia possess no functional chemokine receptor
apoptosis of neurons and cells of the retinal vascular system
exhibit increased neuronal apoptosis, which indicates the
in a retinal cell culture by the application of antibodies against
neuroprotective role of microglia.197,198 Apart from the
a-A- and a-B-crystallin, as well as against HSP27.153 This,
supportive and protective function of the glia, an injury
however, is not directly comparable with the induction of a
caused by degenerative processes or glial activation in
complex autoimmune disease in vivo.
glaucomatous eyes does damage.32,187,199 At least in the eye,
In the case of paraneoplastic neurologic disease (PND),
the activation correlates with the accumulation of mitogen-
which can occur as a secondary symptom of a tumor disease,
activated protein kinase within the glial cells.200 Such
epitopes generated by tumor cells in the body correspond to
activation occurs in the CNS as a general response to any
those of neurons in the CNS. This leads to the establishment of
form of injury or illness and is considered a cellular attempt to
a neurodegenerative process within the CNS. The therapeutic
maintain the impaired mechanisms.
reduction of autoantibody titer by plasmapheresis provides no
persistent relief of the clinical course.219 Conversely, it is not
possible to induce a PND in animal models by injecting
THE ROLE OF INNATE IMMUNITY IN THE antibodies from infected individuals. Autoantibodies alone are
MANIFESTATION OF CENTRAL NERVOUS SYSTEM therefore not sufficient to initiate a neurodegenerative
AUTOIMMUNE DISEASES disease.219,220 However, it is conceivable that autoantibodies
directed against ocular structures have the potential to activate
The activation of microglia, an unmistakable sign of inflamma- the complement system via binding C1q of the classical
tory processes within the CNS, is documented in patients with pathway.165,221–223
Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, The discovery of activated T cells and macrophages in the
MS, and ALS.201–203 Found in serum and cerebrospinal fluid of brain parenchyma of neurodegenerative disease patients
these patients are elevated titers of IL-6, IL-1, and TNF-a, confirms the participation of the cellular immune response
chemokines of innate immunity that are potent initiators of the directed against the body’s own neural tissue within the
extrinsic apoptosis of neurons.201–203 Especially for IL-1 and CNS.224–232 The cause of inflammatory processes in the CNS

Downloaded from iovs.arvojournals.org on 02/05/2019


IOVS, March 2013, Vol. 54, No. 3 A Summary of the Current Knowledge 2397

may therefore be T cells and autoantibodies from the periphery In the case of pathogen recognition, TLR will induce the
that find access to the CNS, where they are again reactivated by coordinated activation of transcription factor NF-jB (nuclear
local antigen-presenting cells (APC) or the microglia, or, in the factor kappa-light-chain-enhancer of activated B-cells), together
case of antibodies, bind directly to their neuronal epitopes. with the increased expression of cytokines IL-1b and IL-6,
The reactivation of T cells triggers the release of inflammatory chemokine IL-8, and the costimulatory proteins CD80 and
cytokines and thereby the maintenance of the immune CD86 (also known as B7.1 and B7.2) for targeted regulation of
response. Phagocytes float to the site of inflammation, take innate immunity.225,249 In general, TLR are dimer-organized,
up cell debris by endocytosis, present fragments thereof in mostly outward-directed receptors, activated by pathogen-
peptide size on the major histocompatibility complex (MHC)-II associated molecular patterns (PAMPs). The PAMPS include
on their surface, and thus again activate the adaptive immune substances such as lipopolysaccharide or bacterial CpG DNA,
response. The adaptive immunity leads to antigen-specific which is characteristic for Gram-negative bacteria.249–251 Of
CD4þ T-helper cell activation and differentiation of naive B cells the total of 12 isotypes so far identified, TLR 3, 7, 8, and 9
into antibody-producing plasma cells in the periphery. These monitor the internal cellular compartments.242,252–254
antibodies cross the blood–brain barrier, particularly under In animal models, experimental elevation of IOP leads to
pathologic circumstances, but even at elevated IOP.233,234 increased expression of TLR 2, 3, and 4 and of HSP27, HSP60,
These immunoglobulins not only bind to epitopes on the and HSP72, as well as the characteristic TLR signaling cascade
surface but also enter the cell by receptor-mediated endocy- adapter proteins and kinases. These findings were confirmed
tosis, as in the case of a-HSP27 antibody. In the cytoplasm of by proteomic analysis of glaucomatous donor eyes. This
neurons, these immunoglobulins may cause apoptosis.142,151 supports the hypothesis that TLR contribute to the activation
The resulting cellular debris is taken up by the phagocytes of of the innate immune system in glaucoma. The increased HSP
the CNS, the microglia as antigen, and also peripheral T cells. expression seems to stimulate the immune system even
This process is hosting an adaptive immune response to neural further.255
targets in the CNS. The latter may be due to the cross-reactivity between HSP
Through recurring cytokine release and antigen presenta- and TLR, but it can also be explained by the ability of HSPs to
tion, a self-perpetuating cycle is under way. The result is present antigens.256,257 Thus, TLR 4 reacts on HSP27 and
apoptosis of neurons, increased permeability of the blood– HSP60, and TLR 2 recognizes glutathione S-transferase as
brain barrier, and upregulation of neuronal MHC-I, causing a PAMP.258–260 HSP72 interacts with TLR 2 and TLR 4.261 This
chronic activation of innate immunity and establishment of an capability of HSPs to cause cross-reactivity can be explained by
adaptive autoimmune response in the CNS. The induction of their distinctive antigenicity. Because HSPs are among the most
intercellular adhesion molecule 1 (ICAM 1) and vascular highly conserved and abundant proteins in nature, as a logical
endothelial growth factor (VEGF) by proinflammatory cyto- consequence they are also among the major antigenic proteins
kines of innate and adaptive immunity leads to further for pathogens like bacteria, mycobacteria, and parasites, which
impairment of the barrier function of the blood–brain barrier, share HSP epitopes with human HSPs.169,262,263 Via the
since they allow the passage of activated T lymphocytes235,236 stimulation of innate immunity by a cross-reaction, which is
(Fig. 1). not in these cases caused by molecular mimicry, autoreactive
Even in the absence of inflammatory events in the CNS, the mechanisms are set in place.264–266 Molecular mimicry with a
transfer of T cells in the subarachnoid space (SAS) between pathogen initially includes ‘‘appearance’’ to the surface
blood and cerebrospinal fluid is feasible.237 Undoubtedly the properties of host cells to evade the surveillance of the
permeability of the blood–brain barrier could play a major role immune defense.267,268 Whether this is caused by cross-
in the pathogenesis of neurodegenerative processes in the CNS reactivity observed in phylogenetic conservation or molecular
such as Alzheimer’s disease, MS, and human immunodeficiency mimicry is obviously dependent on the relevant antigen.
virus (HIV)-associated encephalitis.202,238 Recently, Howell et Either way, the consequences are the same. As seen in the
al. documented the leukocyte transendothelial migration cross-reactivity caused by conserved amino acid sequences,
pathway as activated in the DBA/2J glaucoma model. This mimicry may also establish autoreactive processes.269–271 The
results in the inflow of proinflammatory monocytes (and/or correlation between the initiation of aberrant immune
monocyte-derived cells) into the optic nerve prior to responses due to molecular mimicry between pathogen and
detectable neuronal damage.239 host tissue has been unveiled in the development of numerous
autoimmune diseases. The simultaneous occurrence of enteric
bacteria or virus infection and the onset of autoimmune
INDUCTION OF INNATE IMMUNITY AS A PATHOGENIC processes speaks for itself.272,273 For glaucoma, an infection
FACTOR IN THE CNS with Helicobacter pylori is discussed as a disease-causing
trigger.269,270,274
Today, we know that innate immune responses are responsible
for many chronic neurodegenerative processes within the
CNS.236 The retina in this case has its own immune system, WHERE DO THE NEURONAL ANTIGENS IN THE
including immune cells: microglia, dendritic cells, and even PERIPHERY ORIGINATE?
their own perivascular macrophages, all components of the
innate immunity.240 An essential component of innate immu- Besides the already mentioned cross-reactions, other theories
nity is receptor-mediated pathogen recognition by the Toll-like could help to explain the existence of CNS-specific antigens
receptor family (TLR) and triggering receptor expressed on and autoantibodies in the periphery. Proteins, fragmented or
myeloid cells (TREM). These two classes of receptors are found oxidatively modified by reactive oxygen species (ROS) or
on the microglia, neutrophils, dendritic cells, and macrophages metalloproteinases (MMPs), could serve as autoanti-
in the CNS and the periphery.241–246 Since TREM was identified gens.155,156,275,276 Examples include HSP27, HSP70, and
only in 2000, it is still not really clear what their ligands are. HSP90, which are known to be substrates for MMP-9.
Nevertheless it seems that TREM modulates the activity of TLR; Autoantibodies directed against these HSPs exhibit in-
that is, TREM-1 has an activating and TREM-2 an inhibitory creased reactivity in glaucoma and other neurodegenerative
influence on TLR.247,248 diseases such as MS or Guillain-Barré syndrome.153,155,277,278

Downloaded from iovs.arvojournals.org on 02/05/2019


2398 Rieck IOVS, March 2013, Vol. 54, No. 3

FIGURE 1. Hypothetical mechanism of neurodegeneration in the CNS involving innate and adaptive immunity. Damage of peripheral organs leads to
the activation of immunological processes involving antigen-presenting cells (APC). The APC float to local lymph nodes where activation of the
adaptive immune response takes place. (3, 4) The following clonal expansion of B cells leads to the production of antigen-specific antibodies. (5)
These antibodies pass through the permeable blood–brain barrier. (6) In the CNS, the antibodies bind to neuronal antigens, interfering with their
function, and cause cellular stress and finally apoptosis of the neuron. (7) The apoptosis of neurons leads to activation of microglia. (8) Activated
microglia phagocytose the apoptotic cells and present their antigens on their surface. (9) Microglia secrete proinflammatory cytokines. Increased
titers of vascular endothelial growth factor (VEGF) and the intercellular adhesion molecule 1 (ICAM 1) lead to increased permeability of the blood–
brain barrier. (11) This leads to further influx of T lymphocytes. (12, 13) T cells and neuronal antigens activate microglia, which also leads to the
destruction of neurons. (14) Alternatively, cellular damage or pathogens can cause chronic immune responses of the nonadaptive immune defense.
Reprinted with permission from Nguyen MD, Julien JP, Rivest S. Innate immunity: the missing link in neuroprotection and neurodegeneration? Nat
Rev Neurosci. 2002;3:216–227. Copyright 2002 Macmillan Publishers Ltd: Nature Reviews Neuroscience.

THE JANUS-HEADED COMPLEMENT SYSTEM This allows the formation of membrane attack complex
(MAC), the direct elimination of the target cell by a lytic
In addition to the cellular components of immunological process. These are the well-known facts about the function of
surveillance, the complement system represents the main the complement system. But how can one interpret recent
component of the innate immune response that can be findings regarding increased transcription rates and accumula-
activated by the so-called classical path, the lectin pathway, tion of complement protein C1q and downstream complement
and the alternative pathway. protein C3 within the adult glaucomatous retina?280,281
C1q represents the first component of the classical As described above, the production of autoantibodies is a
pathway, which can lead, either via antibody mediation or hallmark of many autoimmune diseases, as well as in
via direct binding, to complement activation. autoimmune glaucoma.7,8,47,161–163,165,172,179,282–292 These au-
In the case of C1q binding to an apoptotic cell, a pathogen, toantibodies may bind antigens on cell surfaces or form
or cell debris, the activation of a protease cascade directs the immune complexes after catching circulating antigens, which
synthesis of complement component C3 (opsonization).279 may, if not rendered harmless in time, accumulate in lymph
Opsonization with C3 fragments C3b and iCb heads the nodes and diverse organs such as the kidney glomeruli. These
elimination of debris by direct activation of C3 receptors on immune complexes have the potential to activate the
macrophages and microglial phagocytosis. Activated C3 has the complement cascade via the classical pathway.171,292,293
ability to activate the terminal components of the complement However, in the OHT animal model (DBA/2 mice) and
system. donor eyes of OHT patients, transcription rates of C1q and C3

Downloaded from iovs.arvojournals.org on 02/05/2019


IOVS, March 2013, Vol. 54, No. 3 A Summary of the Current Knowledge 2399

FIGURE 2. Distribution of C1q transcripts in the retina. Compared to the control (A) strong signals for C1q can be detected in OHT patients in the
ganglion cell layer (B) and the optic nerve head (ONH, [C]) strong signals for C1q (arrow). Reprinted with permission from Kuehn MH, Kim CY,
Ostojic J, et al. Retinal synthesis and deposition of complement components induced by ocular hypertension. Exp Eye Res. 2006;83:620–628.
Copyright 2006 Elsevier.

are significantly increased. Twenty-eight days after experimen- the blood–brain barrier.209,233,234,310,311 The reactivation of the
tal OHT induction, an accumulation of C1q and C3 transcripts complement cascade is noticeable by an increased transcrip-
is detected in the RGC layer and the ONH29 (Fig. 2). tion rate of C1q, C3, and C4 and the subsequent accumulation
This expression of C1q and C3 correlates with the of these proteins within the inner plexiform layer
activation of glial cells by the complement system. The (IPL).29,280,281,304,312 Synapses, ‘‘tagged’’ with C1q and/or
activated glia hereupon responds with the secretion of NO, C3b, are phagocytized by microglia; downstream formation
TNF-a, superoxide (O2–), and other effector substances. This of the MAC results in synapse loss without participation of glial
involves glia in the reorganization of the ONH and the cells. Apart from the neurotoxic effects of a misdirected
destruction of RGCs.56,294–297 Additionally, the classical com- complement cascade, the decease of RGCs is thought to be
plement cascade contributes to development and function caused by proapoptotic TNF-a and the excitotoxic effect of
refinement of the CNS and visual system via pruning of glutamate, which is supported through the downregulation of
inappropriate synapses.281,298–300 This process of coordinated glutamate transporters during reactive gliosis. What’s left of the
synapse elimination may be facilitated through the ability of RGCs after their demise is thought to be opsonized and
developing CNS neurons to bind C1q directly. This allows removed by the complement system.299 The ongoing elimina-
activation of the classical complement cascade independently tion of apoptotic bodies, cells, and debris has a high priority,
of antibodies.301 However, the involvement of the complement because in the case of aggregation they endanger the
system in synapse elimination is strictly limited to the metabolism and offer an autologous immune epitope.313
development phase of the CNS and visual system under Where the C1q-controlled neutralization of endogenous
normal conditions, but seems to be reactivated in glaucoma. antigens is defective, a misallocation of the immune system
Recent studies point in the direction that components of the may result in the establishment of autoimmune processes such
adaptive immune system also participate in the developmental as SLE or glomerulonephritis.314
synaptic refinement. In particular, class I major histocompa- Generally, it appears that a deviation of the complement
tibily complex (MHC-I), which is not expressed by neurons system is responsible for a number of neurodegenerative
under physiological conditions, is detectable on developing processes, such as ischemic conditions within the CNS, MS,
neurons and on neurons under inflammatory conditions.302,303 Alzheimer’s, and Parkinson’s disease.315–318 Whereas C1q has a
It is intriguing to speculate that components of the comple- role as an eliminator of superfluous synaptic connections
ment pathway may be interacting with MHC-I, mediating during a short phase of embryonic and postnatal maturation of
synapse elimination. To date, such an interaction is elusive. the CNS, its expression in the adult CNS is suppressed under
However, the expression of the MHC-I complex on neuronal physiological conditions. However, the expression of C1q by
surfaces makes axons vulnerable to attacks of CD8þ T cells by neurons and microglia under ischemic, traumatic, and other
perforin-mediated cytotoxicity and the release of granzyme227 pathogenic influences is raised very quickly. Apart from
(Fig. 3). glaucoma, the massive upregulation of C1q expression is also
Reactive gliosis, release of inflammatory cytokines, and detectable in Alzheimer’s disease and ALS.32,304,312,319–323
upregulation of the complement cascade as hallmarks of The key to understanding these degenerative events is to be
developmental synapse elimination in early stages of glaucoma found in the extensive cross talk between complement and
reoccur in many disease models, as well as in eyes of human TLR signaling pathways. This complement–TLR interplay,
donors.29,280,281,304 Recent work investigating the first steps of based on synergistic or antagonistic interactions, reinforces
glaucoma genesis in the DBA/2J mouse points to the activation innate immunity or regulates excessive inflammation. The
of astrocytes as one of the earliest events in glaucoma interaction of complement on TLR signaling is primarily
pathogenesis. This awakening of the innate immunity seems executed via the C5a receptor (C5aR), and to a much lesser
to be triggered by a contusion of RGC axons traversing the extent via the C3a receptor (C3aR). This cross talk appears to
sclera through the lamina cribrosa.10,126,305 Since the lamina is involve the mitogen-activated protein kinase (MAPK) extracel-
the weakest part of the sclera, elevated IOP inevitably leads to lular signal-regulated kinase (ERK1/2), and the c-Jun N-terminal
mechanical stress on RGC axons at this point.2,87,305,306 In the kinase (JNK).324 While the mutual reinforcement of comple-
course of glaucoma progression, microglial proliferation and ment cascade and TLR with its increased TNF-a, IL-1b, and IL-6
activation are accompanied by the secretion of TNF-a and IL- responses might be useful for rapid infection fighting, such an
1b.57,210,295,307–309 As a consequence, this release of inflam- enhanced inflammatory response can slip out of control.
matory cytokines have the potential to impair the integrity of Several membrane-associated proteins should help to control

Downloaded from iovs.arvojournals.org on 02/05/2019


2400 Rieck IOVS, March 2013, Vol. 54, No. 3

FIGURE 3. Cytotoxic T cells use three different routes to destroy their target. Initially the CD8þ T cell recognizes the target via the interaction of its
T-cell receptor (TCR) with the target cell’s MHC-I complex, including presented peptide (square). The destruction of the target cell is performed by
either (i) release of cytotoxic granules and the resulting perforation of the cell membrane, (ii) activation of the target cell’s Fas/CD95 receptor by
Fas ligand/CD95 ligand, or (iii) the release of cytokines such as TNF-a. Reprinted with permission from Neumann H, Medana IM, Bauer J, Lassmann
H. Cytotoxic T lymphocytes in autoimmune and degenerative CNS diseases. Trends Neurosci. 2002;25:313–319. Copyright 2002 Elsevier.

complement action to prevent the host from unintended from patient to patient with regard to the course of disease, the
injury. These proteins are decay-accelerating factor (DAF), severity, and the underlying dysfunction of the immune system.
membrane cofactor protein (MCP), complement receptor type To minimize these uncertainties, the creation of a person-
1 (CR1), and CD59. Although the complement cascade is alized ‘‘autoantibody signature’’ is proposed by some authors.
accepted as participating in autoimmunity, little is known Using the antigen microarray technique, this tool would
concerning the degree to which membrane complement provide insight into the course of disease, particularly with
regulatory proteins are capable of modulating complement reference to the patient’s response to a therapeutic agent.172 A
action. In any case, knockout mice lacking these regulatory recent study, performed with patients suffering from rheuma-
proteins display a higher susceptibility to autoimmune injury toid arthritis (RA), investigated the antibody profile in response
than wild-type mice.325
to the anti-TNF therapeutic agent etanercept. In summary, a
Increasingly, the complement system, traditionally known
panel of proteomic markers was shown to be able to
as a peripheral complemental function, migrates to the center
of the immune system as a global mediator of immune distinguish responders from nonresponders.291 Suggesting that
surveillance, cell homeostasis, and tissue development and TNF-a may also be an important factor in the neurodegener-
repair. The review article by Ricklin et al.293 offers additional ative process of glaucoma, a OHT mouse model was used to
insights into recent findings concerning the complement investigate the effect of etanercept on glaucoma. In fact, the
system. therapeutic anti-TNF-a antibody etanercept is effective in
rescuing RGCs from OHT-induced death.329 An investigation
of the neuroprotective effect of etanercept could enable the
CLINICAL DIAGNOSIS OF AUTOIMMUNE GLAUCOMA discovery of proteomic factors that are involved in the
pathogenesis of glaucoma. A panel of proteomic markers that
To diagnose glaucoma caused by autoimmunity, the attending allows the distinction of healthy from those suffering from
physician has no choice but first to exclude all other causes of
glaucoma is an enticing perspective.
glaucoma. These include elevated IOP in high-pressure
glaucoma. In the case of NTG, pathogenic factors like ischemia, While the investigation of autoantibody profiles as a
migraine, systemic nocturnal hypotension, or sleep apnea must methodical approach might help to increase understanding
be excluded as a cause for glaucoma. In summary, it can be said of the mechanisms of aberrant immunity in glaucoma, the
that the diagnosis of autoimmune glaucoma is a diagnosis of identification of diagnostic biomarkers would be a great
exclusion.326 benefit. Proteomic biomarkers are established tools in early
Based on the available data on autoreactivities, one can diagnosis and are well suited to monitoring the course of
presume a tissue specificity of the immune response in numerous diseases. As an example, superoxide dismutase is
glaucoma. This specificity derives from the autoantigen that used in prenatal diagnostics as a biomarker for Down
has been initially recognized.327,328 A complicating factor is syndrome because of the excessive expression of genes
that any given autoimmune disease is heterologous, varying encoded on chromosome 21.330,331

Downloaded from iovs.arvojournals.org on 02/05/2019


IOVS, March 2013, Vol. 54, No. 3 A Summary of the Current Knowledge 2401

In glaucoma, distinct immunglobulins, such as autoantibod- 5. Henderson PA, Medeiros FA, Zangwill LM, Weinreb RN.
ies to alpha-fodrin163 or glutathione S-transferase,165 may serve Relationship between central corneal thickness and retinal
as biomarkers as well as proteins that are significantly linked to nerve fiber layer thickness in ocular hypertensive patients.
cytotoxic conditions in glaucoma. Actually, transthyretin has Ophthalmology. 2005;112:251–256.
been identified as a biomarker for glaucoma in aqueous humor 6. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological
of patients with POAG.332 Another biomarker for POAG, brain- phenomenon with wide-ranging implications in tissue
derived neurotrophic factor (BDNF), could be detected in the kinetics. Br J Cancer. 1972;26:239–257.
sera of glaucoma patients.333 7. Maruyama I, Ikeda Y, Nakazawa M, Ohguro H. Clinical roles of
Markers that correspond with oxidative stress are superox- serum autoantibody against neuron-specific enolase in
ide dismutase,334–339 glutathione peroxidase,334,337,340 ascor- glaucoma patients. Tohoku J Exp Med. 2002;197:125–132.
bic acid,334,335,340 a-tocopherol,340,341 retinol,228,335 nitric 8. Maruyama I, Ohguro H, Ikeda Y. Retinal ganglion cells
oxide,43,57,157,210,221,295,296,337,342–346 8-hydroxy-20-deoxygua- recognized by serum autoantibody against gamma-enolase
nosine,347 and malondialdehyde.337,348 Detailed information found in glaucoma patients. Invest Ophthalmol Vis Sci. 2000;
concerning all glaucoma biomarkers identified to date is 41:1657–1665.
provided in the review article of Pinazo-Duran et al.349 9. Neufeld AH, Sawada A, Becker B. Inhibition of nitric-oxide
However, as of now, no neuroprotective treatments that synthase 2 by aminoguanidine provides neuroprotection of
directly target pathogenic mechanisms in the retina or optic retinal ganglion cells in a rat model of chronic glaucoma.
nerve and no laboratory diagnostics are approved for clinical Proc Natl Acad Sci U S A. 1999;96:9944–9948.
use against glaucoma. But the progress that glaucoma research 10. Nickells RW, Howell GR, Soto I, John SW. Under pressure:
has made in recent years gives reason to hope that this will cellular and molecular responses during glaucoma, a
change soon. common neurodegeneration with axonopathy. Annu Rev
Neurosci. 2012;35:153–179.
11. Quigley HA. Ganglion cell death in glaucoma: pathology
SUMMARY recapitulates ontogeny. Aust N Z J Ophthalmol. 1995;23:85–
According to current knowledge, a sole trigger for glaucoma 91.
can be excluded. Apparently the lamina cribrosa as the most 12. Quigley HA. Neuronal death in glaucoma. Prog Retin Eye
vulnerable site of the RGCs is of special interest. This may be Res. 1999;18:39–57.
the position at which impact-induced stress triggers the 13. Quigley HA. Glaucoma. Lancet. 2011;377:1367–1377.
activation of one or more self-destruct programs, depending 14. Weinreb RN, Khaw PT. Primary open-angle glaucoma.
on the severity of the impact, WD, DB, or the initiation of a Lancet. 2004;363:1711–1720.
gliosis—or none of these. While it is now understood that 15. Whitmore AV, Libby RT, John SWM. Glaucoma: thinking in
many cell types and mechanisms respond to cellular stress, it is new ways–a rôle for autonomous axonal self-destruction and
not clear where the earliest pathologic events take place and other compartmentalised processes? Prog Retin Eye Res.
where the key trigger comes from. In this context, the CNS- 2005;24:639–662.
specific immune system is of particular importance. Under 16. Harrington DO. The Bjerrum scotoma. Trans Am Ophthal-
physiological conditions, this ensures a perfect homeostasis. mol Soc. 1964;62:324–348.
However, this state can be destroyed in the next moment 17. Garcia-Valenzuela E, Shareef S, Walsh J, Sharma SC. Pro-
under conditions of cellular stress. As a trigger for cellular grammed cell death of retinal ganglion cells during experi-
stress, a variety of possible factors are under consideration. mental glaucoma. Exp Eye Res. 1995;61:33–44.
Among these factors are reactive oxygen species (ROS),
18. Glovinsky Y, Quigley HA, Dunkelberger GR. Retinal ganglion
protein aggregation, ischemia, vasospasm, deficient neurotro- cell loss is size dependent in experimental glaucoma. Invest
phin supply, and even light. However, the early events in glial Ophthalmol Vis Sci. 1991;32:484–491.
activation seem to be crucial for the manifestation of glaucoma.
19. Nickells RW. Retinal ganglion cell death in glaucoma: the
These events need further investigation, because they may be a
how, the why, and the maybe. J Glaucoma. 1996;5:345–356.
key to an effective therapy.
20. Quigley HA, Nickells RW, Kerrigan LA, Pease ME, Thibault DJ,
Zack DJ. Retinal ganglion cell death in experimental
Acknowledgments glaucoma and after axotomy occurs by apoptosis. Invest
The author thanks Barbara Pullmann, Annette Weber, and Ulrich Ophthalmol Vis Sci. 1995;36:774–786.
Hoeger for valuable support. The author thanks Paul L. Kaufman 21. Bjerrum J. Om en tilføjelse til den sædvanlige synsfelt –
for detailed and helpful comments. undersögelse samt om synsfeltet ved glaukom. Nord ophthal
Tidsskrift. 1889;2:141–185.
References 22. Jay JL, Murray SB. Early trabeculectomy versus conventional
management in primary open angle glaucoma. Br J Oph-
1. Quigley HA, Broman AT. The number of people with thalmol. 1988;72:881–889.
glaucoma worldwide in 2010 and 2020. Br J Ophthalmol. 23. Zur D, Ullman S. Filling-in of retinal scotomas. Vis Res. 2003;
2006;90:262–267. 43:971–982.
2. Burgoyne CF. A biomechanical paradigm for axonal insult 24. Gloor BP, Sarra GM. Visusverlust und Sehstörung (2. Teil).
within the optic nerve head in aging and glaucoma. Exp Eye Schweiz Med Forum. 2004;4:308–312.
Res. 2010;93:120–132. 25. Kass MA, Heuer DK, Higginbotham EJ, et al. The Ocular
3. Fechtner RD, Weinreb RN. Mechanisms of optic nerve Hypertension Treatment Study: a randomized trial deter-
damage in primary open angle glaucoma. Surv Ophthalmol. mines that topical ocular hypotensive medication delays or
1994;39:23–42. prevents the onset of primary open-angle glaucoma. Arch
4. Ferrer E. Trabecular meshwork as a new target for the Ophthalmol. 2002;120:701–713.
treatment of glaucoma. Drug News Perspect. 2006;19:151– 26. Weber AJ, Chen H, Hubbard WC, Kaufman PL. Experimental
158. glaucoma and cell size, density, and number in the primate

Downloaded from iovs.arvojournals.org on 02/05/2019


2402 Rieck IOVS, March 2013, Vol. 54, No. 3

lateral geniculate nucleus. Invest Ophthalmol Vis Sci. 2000; 47. Grus FH, Joachim SC, Pfeiffer N. Analysis of complex
41:1370–1379. autoantibody repertoires by surface-enhanced laser desorp-
27. Gordon MO, Beiser JA, Brandt JD, et al. The Ocular tion/ionization-time of flight mass spectrometry. Proteomics.
Hypertension Treatment Study: baseline factors that predict 2003;3:957–961.
the onset of primary open-angle glaucoma. Arch Ophthal- 48. CNTGSG. Comparison of glaucomatous progression between
mol. 2002;120:714–720. untreated patients with normal-tension glaucoma and pa-
28. Loewen NA, Liu JH, Weinreb RN. Increased 24-hour variation tients with therapeutically reduced intraocular pressures. Am
of human intraocular pressure with short axial length. Invest J Ophthalmol. 1998;126:487–497.
Ophthalmol Vis Sci. 2010;51:933–937. 49. CNTGSG. The effectiveness of intraocular pressure reduction
29. Kuehn MH, Kim CY, Ostojic J, et al. Retinal synthesis and in the treatment of normal-tension glaucoma. Am J Oph-
deposition of complement components induced by ocular thalmol. 1998;126:498–505.
hypertension. Exp Eye Res. 2006;83:620–628. 50. Kuehn MH, Fingert JH, Kwon YH. Retinal ganglion cell death
30. Naskar R, Wissing M, Thanos S. Detection of early neuron in glaucoma: mechanisms and neuroprotective strategies.
degeneration and accompanying microglial responses in the Ophthalmol Clin North Am. 2005;18:383–395, vi.
retina of a rat model of glaucoma. Invest Ophthalmol Vis Sci. 51. Crawford ML, Harwerth RS, Smith EL III, Mills S, Ewing B.
2002;43:2962–2968. Experimental glaucoma in primates: changes in cytochrome
31. Guo L, Moss SE, Alexander RA, Ali RR, Fitzke FW, Cordeiro oxidase blobs in V1 cortex. Invest Ophthalmol Vis Sci. 2001;
MF. Retinal ganglion cell apoptosis in glaucoma is related to 42:358–364.
intraocular pressure and IOP-induced effects on extracellular 52. Yücel YH, Zhang Q, Gupta N, Kaufman PL, Weinreb RN. Loss
matrix. Invest Ophthalmol Vis Sci. 2005;46:175–182. of neurons in magnocellular and parvocellular layers of the
32. Steele MR, Inman DM, Calkins DJ, Horner PJ, Vetter ML. lateral geniculate nucleus in glaucoma. Arch Ophthalmol.
Microarray analysis of retinal gene expression in the DBA/2J 2000;118:378–384.
model of glaucoma. Invest Ophthalmol Vis Sci. 2006;47:977– 53. Yücel YH, Zhang Q, Weinreb RN, Kaufman PL, Gupta N.
985. Atrophy of relay neurons in magno- and parvocellular layers
33. Johnson EC, Deppmeier LM, Wentien SK, Hsu I, Morrison JC. in the lateral geniculate nucleus in experimental glaucoma.
Invest Ophthalmol Vis Sci. 2001;42:3216–3222.
Chronology of optic nerve head and retinal responses to
elevated intraocular pressure. Invest Ophthalmol Vis Sci. 54. Yücel YH, Zhang Q, Weinreb RN, Kaufman PL, Gupta N.
2000;41:431–442. Effects of retinal ganglion cell loss on magno-, parvo-,
koniocellular pathways in the lateral geniculate nucleus and
34. Grehn F, Hollo G, Lachkar Y, Migdal C, Thygesen J.
visual cortex in glaucoma. Prog Retin Eye Res. 2003;22:465–
Terminology and Guidelines for Glaucoma. Vol. 2. Savona,
481.
Italy: European Glaucoma Society; 2003.
55. Fukuchi T, Ueda J, Hanyu T, Abe H, Sawaguchi S. Distribution
35. Weinreb RN, Levin LA. Is neuroprotection a viable therapy
and expression of transforming growth factor-beta and
for glaucoma? Arch Ophthalmol. 1999;117:1540–1544.
platelet-derived growth factor in the normal and glaucoma-
36. Nickells RW. From ocular hypertension to ganglion cell tous monkey optic nerve heads. Jpn J Ophthalmol. 2001;45:
death: a theoretical sequence of events leading to glaucoma. 592–599.
Can J Ophthalmol. 2007;42:278–287.
56. Hernandez MR. The optic nerve head in glaucoma: role of
37. Coleman AL. Glaucoma. Lancet. 1999;354:1803–1810. astrocytes in tissue remodeling. Prog Retin Eye Res. 2000;19:
38. Mackenzie P, Cioffi G. How does lowering of intraocular 297–321.
pressure protect the optic nerve? Surv Ophthalmol. 2008; 57. Zhong YS, Leung CK, Pang CP. Glial cells and glaucomatous
53(suppl 1):S39–S43. neuropathy. Chin Med J (Engl). 2007;120:326–335.
39. Mozaffarieh M, Flammer J. Is there more to glaucoma 58. Nickells RW. Apoptosis of retinal ganglion cells in glaucoma:
treatment than lowering IOP? Surv Ophthalmol. 2007; an update of the molecular pathways involved in cell death.
52(suppl 1):S174–S179. Surv Ophthalmol. 1999;43(suppl 1):S151–S161.
40. Enyedi LB, Freedman SF. Safety and efficacy of brimonidine in 59. Calandrella N, Scarsella G, Pescosolido N, Risuleo G.
children with glaucoma. J Pediatr Ophthalmol Strabismus. Degenerative and apoptotic events at retinal and optic
2001;5:281–284. nerve level after experimental induction of ocular hyper-
41. Schuettauf F, Quinto K, Naskar R, Zurakowski D. Effects of tension. Mol Cell Biochem. 2007;301:155–163.
anti-glaucoma medications on ganglion cell survival: the 60. Levkovitch-Verbin H, Dardik R, Vander S, Melamed S.
DBA/2J mouse model. Vis Res. 2002;42:2333–2337. Mechanism of retinal ganglion cells death in secondary
42. Pfeiffer N, Grierson I, Goldsmith H, Hochgesand D, Winkgen- degeneration of the optic nerve. Exp Eye Res. 2010;91:127–
Bohres A, Appleton P. Histological effects in the iris after 3 134.
months of latanoprost therapy: the Mainz 1 Study. Arch 61. Libby RT, Li T, Savinova OV, et al. Susceptibility to
Ophthalmol. 2001;119:191–196. neurodegeneration in a glaucoma is modified by Bax gene
43. Chidlow G, Wood JP, Casson RJ. Pharmacological neuropro- dosage. PLoS Genet. 2005;1:17–26.
tection for glaucoma. Drugs. 2007;67:725–759. 62. Murakami A, Okisaka S. Neuronal cell death mechanism in
44. Pfeiffer N. Glaukom und okuläre Hypertension - Grund- glaucomatous optic neuropathy. Nippon Ganka Gakkai
lagen, Diagnostik, Therapie. Vol. 1. Stuttgart, Germany: Zasshi. 1998;102:645–653.
Thieme-Verlag; 2001. 63. Nickells RW. The molecular biology of retinal ganglion cell
45. Aung T, Ocaka L, Ebenezer ND, et al. A major marker for death: caveats and controversies. Brain Res Bull. 2004;62:
normal tension glaucoma: association with polymorphisms in 439–446.
the OPA1 gene. Hum Genet. 2002;110:52–56. 64. Tatton WG, Olanow CW. Apoptosis in neurodegenerative
46. Bonomi L, Marchini G, Marraffa M, et al. Prevalence of diseases: the role of mitochondria. Biochim Biophys Acta.
glaucoma and intraocular pressure distribution in a defined 1999;1410:195–213.
population: the Egna-Neumarkt study. Ophthalmology. 1998; 65. Pease ME, McKinnon SJ, Quigley HA, Kerrigan-Baumrind LA,
105:209–215. Zack DJ. Obstructed axonal transport of BDNF and its

Downloaded from iovs.arvojournals.org on 02/05/2019


IOVS, March 2013, Vol. 54, No. 3 A Summary of the Current Knowledge 2403

receptor TrkB in experimental glaucoma. Invest Ophthalmol 85. Valdez G, Philippidou P, Rosenbaum J, Akmentin W, Shao Y,
Vis Sci. 2000;41:764–774. Halegoua S. Trk-signaling endosomes are generated by Rac-
66. Agar A, Li S, Agarwal N, Coroneo MT, Hill MA. Retinal dependent macroendocytosis. Proc Natl Acad Sci U S A.
ganglion cell line apoptosis induced by hydrostatic pressure. 2007;104:12270–12275.
Brain Res. 2006;1086:191–200. 86. Allcutt D, Berry M, Sievers J. A quantitative comparison of the
67. Agar A, Yip SS, Hill MA, Coroneo MT. Pressure related reactions of retinal ganglion cells to optic nerve crush in
apoptosis in neuronal cell lines. J Neurosci Res. 2000;60: neonatal and adult mice. Brain Res. 1984;16:219–230.
495–503. 87. Morgan JE, Jeffery G, Foss AJE. Axon deviation in the human
68. Schlamp CL, Li Y, Dietz JA, Janssen KT, Nickells RW. lamina cribrosa. Br J Ophthalmol. 1998;82:680–683.
Progressive ganglion cell loss and optic nerve degeneration 88. Radius RL, Anderson DR. Rapid axonal transport in primate
in DBA/2J mice is variable and asymmetric. BMC Neurosci. optic nerve. Distribution of pressure-induced interruption.
2006;7:66. Arch Ophthalmol. 1981;99:650–654.
69. Leske MC. The epidemiology of open angle glaucoma: a 89. Dandona L, Hendrickson A, Quigley HA. Selective effects of
review. Am J Epidemiol. 1983;118:166–191. experimental glaucoma on axonal transport by retinal
70. Pfeiffer N. Results of the ‘‘Ocular Hypertension treatment ganglion cells to the dorsal lateral geniculate nucleus. Invest
study.’’ Ophthalmologe. 2005;102:230–234. Ophthalmol Vis Sci. 1991;32:1593–1599.
71. Minckler DS, Bunt AH, Johanson GW. Orthograde and 90. Bellezza AJ, Rintalan CJ, Thompson HW, Downs JC, Hart RT,
retrograde axoplasmic transport during acute ocular hyper- Burgoyne CF. Deformation of the lamina cribrosa and anterior
tension in the monkey. Invest Ophthalmol Vis Sci. 1977;16: scleral canal wall in early experimental glaucoma. Invest
426–441. Ophthalmol Vis Sci. 2003;44:623–637.
72. Quigley HA, McKinnon SJ, Zack DJ, et al. Retrograde axonal 91. Pena JD, Agapova O, Gabelt BT, et al. Increased elastin
transport of BDNF in retinal ganglion cells is blocked by expression in astrocytes of the lamina cribrosa in response to
acute IOP elevation in rats. Invest Ophthalmol Vis Sci. 2000; elevated intraocular pressure. Invest Ophthalmol Vis Sci.
41:3460–3466. 2001;42:2303–2314.
73. Mansour-Robaey S. Effects of ocular injury and administration 92. Jonas JB. Role of cerebrospinal fluid pressure in the pathogen-
of brain-derived neurotrophic factor on survival and esis of glaucoma. Acta Ophthalmol. 2011;89:505–514.
regrowth of axotomized retinal ganglion cells. Proc Natl 93. Li Y, Schlamp CL, Poulsen GL, Jackson MW, Griep AE,
Acad Sci U S A. 1994;91:1632–1636. Nickells RW. p53 regulates apoptotic retinal ganglion cell
74. Sawai H, Clarke DB, Kittlerova P, Bray GM, Aguayo AJ. Brain- death induced by N-methyl-D-aspartate. Mol Vis. 2002;8:341–
derived neurotrophic factor and neurotrophin-4/5 stimulate 350.
growth of axonal branches from regenerating retinal ganglion 94. Li Y, Schlamp CL, Poulsen KP, Nickells RW. Bax-dependent
cells. J Neurosci. 1996;16:3887–3894. and independent pathways of retinal ganglion cell death
75. Klöcker N, Bräunling F, Isenmann S, Bähr M. In vivo induced by different damaging stimuli. Exp Eye Res. 2000;71:
neurotrophic effects of GDNF on axotomized retinal ganglion 209–213.
cells. Neuroreport. 1997;8:3439–3442. 95. Wax MB, Tezel G, Yang J, et al. Induced autoimmunity to heat
76. Ji JZ, Elyaman W, Yip HK, et al. CNTF promotes survival of shock proteins elicits glaucomatous loss of retinal ganglion
retinal ganglion cells after induction of ocular hypertension cell neurons via activated T-cell-derived Fas-ligand. J Neuro-
in rats: the possible involvement of STAT3 pathway. Eur J sci. 2008;28:12085–12096.
Neurosci. 2004;19:265–272. 96. McKinnon SJ. The cell and molecular biology of glaucoma:
77. Zweifel LS, Kuruvilla R, Ginty DD. Functions and mechanisms common neurodegenerative pathways and relevance to
of retrograde neurotrophin signalling. Nat Rev Neurosci. glaucoma. Invest Ophthalmol Vis Sci. 2012;53:2485–2487.
2005;6:615–625. 97. Yaar M, Pilch PF, Doyle SM, Eisnehauer PB, Fine RE, Gilchrest
78. Meyer-Franke A, Kaplan MR, Pfrieger FW, Barres BA. GA. Binding of beta-amyloid to the p75 neurotrophin
Characterization of the signaling interactions that promote receptor induces apoptosis. A possible mechanism for
the survival and growth of developing retinal ganglion cells Alzheimer’s disease. J Clin Invest. 1997;100:2333–2340.
in culture. Neuron. 1995;15:805–819. 98. Hafezi F, Marti A, Munz K, Remé CE. Light-induced apoptosis:
79. Weibel D, Kreutzberg GW, Schwab ME. Brain-derived differential timing in the retina and pigment epithelium. Exp
neurotrophic factor (BDNF) prevents lesion-induced axonal Eye Res. 1997;64:963–970.
die-back in young rat optic nerve. Brain Res. 1995;679:249– 99. Harada T, Harada C, Nakayama N, et al. Modification of glial
254. neuronal cell interactions prevents photoreceptor apoptosis
80. Eathiraj S, Pan X, Ritacco C, Lambright DG. Structural basis of during light-induced retinal degeneration. Neuron. 2000;26:
family-wide Rab GTPase recognition by rabenosyn-5. Nature. 533–541.
2005;436:415–419. 100. Lipton SA. Possible role for memantine in protecting retinal
81. Nielsen E, Christoforidis S, Uttenweiler-Joseph S, et al. ganglion cells from glaucomatous damage. Surv Ophthalmol.
Rabenosyn-5, a novel Rab5 effector, is complexed with 2003;48(suppl 1):S38–S46.
Hvps45 and recruited to endosomes through a Fyve finger 101. Schori H, Kipnis J, Yoles E, et al. Vaccination for protection of
domain. J Cell Biol. 2000;151:601–612. retinal ganglion cells against death from glutamate cytotox-
82. Nielsen E, Severin F, Backer JM, Hyman AA, Zerial M. Rab5 icity and ocular hypertension: implications for glaucoma.
regulates motility of early endosomes on microtubules. Nat Proc Natl Acad Sci U S A. 2001;98:3398–3403.
Cell Biol. 1999;1:376–382. 102. Vorwerk CK, Naskar R, Schuettauf F, et al. Depression of
83. Schnatwinkel C, Christoforidis S, Lindsay MR, et al. The Rab5 retinal glutamate transporter function leads to elevated
effector rabankyrin-5 regulates and coordinates different intravitreal glutamate levels and ganglion cell death. Invest
endocytic mechanisms. PLoS Biol. 2004;2:e261. Ophthalmol Vis Sci. 2000;41:3615–3621.
84. Shin H-W, Hayashi M, Christoforidis S, et al. An enzymatic 103. Honkanen RA, Baruah S, Zimmerman MB, et al. Vitreous
cascade of Rab5 effectors regulates phosphoinositide turn- amino acid concentrations in patients with glaucoma
over in the endocytic pathway. J Cell Biol. 2005;170:607– undergoing vitrectomy. Arch Ophthalmol. 2003;121:183–
618. 188.

Downloaded from iovs.arvojournals.org on 02/05/2019


2404 Rieck IOVS, March 2013, Vol. 54, No. 3

104. Ullian EM, Barkis WB, Chen S, Diamond JS, Barres BA. 126. Howell GR, Soto I, Libby RT, John SW. Intrinsic axonal
Invulnerability of retinal ganglion cells to NMDA excitotox- degeneration pathways are critical for glaucomatous damage
icity. Mol Cell Neurosci. 2004;26:544–557. [published online ahead of print January 18, 2012]. Exp
105. Bringmann A, Reichenbach A. Role of Muller cells in retinal Neurol. doi:10.1016/j.expneurol.2012.01.014.
degenerations. Front Biosci. 2001;6:E72–E92. 127. Li Y, Semaan SJ, Schlamp CL, Nickells RW. Dominant
106. Russo R, Rotiroti D, Tassorelli C, et al. Identification of novel inheritance of retinal ganglion cell resistance to optic nerve
pharmacological targets to minimize excitotoxic retinal crush in mice. BMC Neurosci. 2007;8:19.
damage. Int Rev Neurobiol. 2009;85:407–423. 128. Schwartz M. Vaccination for glaucoma: dream or reality?
107. Inoue-Matsuhisa E, Sogo S, Mizota A, Taniai M, Takenaka H, Brain Res Bull. 2004;62:481–484.
Mano T. Effect of MCI-9042, a 5-HT2 receptor antagonist, on 129. Ohlsson M, Bellander BM, Langmoen IA, Svensson M.
retinal ganglion cell death and retinal ischemia. Exp Eye Res. Complement activation following optic nerve crush in the
2003;76:445–452. adult rat. J Neurotrauma. 2003;20:895–904.
108. Bucolo C, Leggio GM, Maltese A, Castorina A, D’Agata V, 130. Tezel G, Yang X, Yang J, Wax MB. Role of tumor necrosis
Drago F. Dopamine-3 receptor modulates intraocular pres- factor receptor-1 in the death of retinal ganglion cells
sure: implications for glaucoma. Biochem Pharmacol. 2011; following optic nerve crush injury in mice. Brain Res.
83:680–686. 2004;996:202–212.
109. Lingor P, Koeberle P, Kügler S, Bähr M. Down-regulation of 131. Waller A. Experiments on the section of the glossopharyngeal
apoptosis mediators by RNAi inhibits axotomy-induced and hypoglossal nerves of the frog, and observations of the
retinal ganglion cell death in vivo. Brain. 2005;128:550–558. alterations produced thereby in the structure of their
110. Osborne NN, Lascaratos G, Bron AJ, Chidlow G, Wood JP. A primitive fibres. Philos Trans R Soc Lond. 1850;140:423–429.
hypothesis to suggest that light is a risk factor in glaucoma 132. Tse MT. Axon degeneration: a new pathway emerges. Nat
and the mitochondrial optic neuropathies. Br J Ophthalmol. Rev Neurosci. 2012;13:516.
2006;90:237–241. 133. Beirowski B, Adalbert R, Wagner D, et al. The progressive
111. Aronica E, Gorter JA, Ijlst-Keizers H, et al. Expression and nature of Wallerian degeneration in wild-type and slow
functional role of mGluR3 and mGluR5 in human astrocytes Wallerian degeneration (WldS) nerves. BMC Neurosis. 2005;
and glioma cells: opposite regulation of glutamate transport- 6:6.
er proteins. Eur J Neurosci. 2003;17:2106–2118. 134. de Lima S, Koriyama Y, Kurimoto T, et al. Full-length axon
112. Naskar R, Vorwerk CK, Dreyer EB. Concurrent downregula- regeneration in the adult mouse optic nerve and partial
tion of a glutamate transporter and receptor in glaucoma. recovery of simple visual behaviors. Proc Natl Acad Sci
Invest Ophthalmol Vis Sci. 2000;41:1940–1944. U S A. 2012;109:9149–9154.
113. Bovolenta P, Wandosell F, Nieto-Sampedro M. CNS glial scar 135. Vidal-Sanz M, Aviles-Trigueros M, Whiteley SJ, Sauve Y, Lund
tissue: a source of molecules which inhibit central neurite RD. Reinnervation of the pretectum in adult rats by
outgrowth. Prog Brain Res. 1992;94:367–379. regenerated retinal ganglion cell axons: anatomical and
functional studies. Prog Brain Res. 2002;137:443–452.
114. Campenot RB. Local control of neurite development by nerve
growth factor. Proc Natl Acad U S A. 1977;74:4516–4519. 136. Sofroniew M, Vinters H. Astrocytes: biology and pathology.
Acta Neuropathol. 2010;119:7–35.
115. Campenot RB. Local control of neurite sprouting in cultured
sympathetic neurons by nerve growth factor. Dev Brain Res. 137. Wax MB, Tezel G, Saito I, et al. Anti-Ro/SS-a positivity and
1987;37:293–301. heat shock protein antibodies in patients with normal-
pressure glaucoma. Am J Ophthalmol. 1998;125:145–157.
116. Castaño A, Bell MD, Perry VH. Unusual aspects of inflamma-
tion in the nervous system: Wallerian degeneration. Neuro- 138. Feder ME, Hofmann GE. Heat-shock proteins, molecular
biol Aging. 1996;17:745–751. chaperones, and the stress response: evolutionary and
ecological physiology. Annu Rev Physiol. 1999;61:243–282.
117. Fu QL, Li X, Shi J, et al. Synaptic degeneration of retinal
ganglion cells in a rat ocular hypertension glaucoma model. 139. Currie RW, Ellison JA, White RF, Feuerstein GZ, Wang X,
Cell Mol Neurobiol. 2009;29:575–581. Barone FC. Benign focal ischemic preconditioning induces
neuronal Hsp70 and prolonged astrogliosis with expression
118. Gillingwater TH, Ribchester RR. The relationship of neuro- of Hsp27. Brain Res. 2000;863:169–181.
muscular synapse elimination to synaptic degeneration and
pathology: insights from WldS and other mutant mice. J 140. Manzerra P, Rush SJ, Brown IR. Temporal and spatial
Neurocytol. 2003;32:863–881. distribution of heat shock mRNA and protein (hsp70) in
the rabbit cerebellum in response to hyperthermia. J
119. Hilliard MA. Axonal degeneration and regeneration: a Neurosci Res. 1993;36:480–490.
mechanistic tug-of-war. J Neurochem. 2009;108:23–32.
141. Franklin TB, Krueger-Naug AM, Clarke DB, Arrigo AP, Currie
120. Jellinger K. Recent advances in our understanding of RW. The role of heat shock proteins Hsp70 and Hsp27 in
neurodegeneration. J Neural Transm. 2009;116:1111–1162. cellular protection of the central nervous system. Int J
121. Linker RA, Sendtner M, Gold R. Mechanisms of axonal Hyperthermia. 2005;21:379–392.
degeneration in EAE–lessons from CNTF and MHC I 142. Tezel G, Wax MB. The mechanisms of hsp27 antibody-
knockout mice. J Neurol Sci. 2005;233:167–172. mediated apoptosis in retinal neuronal cells. J Neurosci.
122. Profyris C, Cheema SS, Zang D, Azari MF, Boyle K, Petratos S. 2000;20:3552–3562.
Degenerative and regenerative mechanisms governing spinal 143. Krueger-Naug AM, Emsley JG, Myers TL, Currie RW, Clarke
cord injury. Neurobiol Dis. 2004;15:415–436. DB. Injury to retinal ganglion cells induces expression of the
123. Yoles E, Schwartz M. Degeneration of spared axons following small heat shock protein Hsp27 in the rat visual system.
partial white matter lesion: implications for optic nerve Neuroscience. 2002;110:653–665.
neuropathies. Exp Neurol. 1998;153:1–7. 144. Garrido C, Bruey JM, Fromentin A, Hammann A, Arrigo AP,
124. Coleman MP, Freeman MR. Wallerian degeneration, WldS, Solary E. HSP27 inhibits cytochrome c-dependent activation
and Nmnat. Annu Rev Neurosci. 2010;33:245–267. of procaspase-9. FASEB J. 1999;13:2061–2070.
125. Kisiswa L, Dervan AG, Albon J, Morgan JE, Wride MA. Retinal 145. Garrido C, Gurbuxani S, Ravagnan L, Kroemer G. Heat shock
ganglion cell death postponed: giving apoptosis a break? proteins: endogenous modulators of apoptotic cell death.
Ophthalmic Res. 2010;43:61–78. Biochem Biophys Res Commun. 2001;286:433–442.

Downloaded from iovs.arvojournals.org on 02/05/2019


IOVS, March 2013, Vol. 54, No. 3 A Summary of the Current Knowledge 2405

146. Tezel G, Hernandez MR, Wax MB. Immunostaining of heat 165. Yang J, Tezel G, Patil RV, Romano C, Wax MB. Serum
shock proteins in the retina and optic nerve head of normal autoantibody against glutathione S-transferase in patients
and glaucomatous eyes. Arch Ophthalmol. 2000;118:511– with glaucoma. Invest Ophthalmol Vis Sci. 2001;42:1273–
518. 1276.
147. Kretz A, Schmeer C, Tausch S, Isenmann S. Simvastatin 166. Yano T, Yamada K, Kimura A, et al. Autoimmunity against
promotes heat shock protein 27 expression and Akt neurofilament protein and its possible association with HLA-
activation in the rat retina and protects axotomized retinal DRB1*1502 allele in glaucoma. Immunol Lett. 2005;100:
ganglion cells in vivo. Neurobiol Dis. 2006;21:421–430. 164–169.
148. Whitlock NA, Agarwal N, Ma JX, Crosson CE. Hsp27 167. Thatcher TH, Gorovsky MA. Phylogenetic analysis of the core
upregulation by HIF-1 signaling offers protection against histones H2A, H2B, H3, and H4. Nucleic Acids Res. 1994;22:
retinal ischemia in rats. Invest Ophthalmol Vis Sci. 2005;46: 174–149.
1092–1098. 168. Garsia RJ, Hellqvist L, Booth RJ, et al. Homology of the 70-
149. Whitlock NA, Lindsey K, Agarwal N, Crosson CE, Ma JX. Heat kilodalton antigens from Mycobacterium leprae and Myco-
shock protein 27 delays Ca2þ-induced cell death in a caspase- bacterium bovis with the Mycobacterium tuberculosis 71-
dependent and -independent manner in rat retinal ganglion kilodalton antigen and with the conserved heat shock
cells. Invest Ophthalmol Vis Sci. 2005;46:1085–1091. protein 70 of eucaryotes. Infect Immun. 1989;57:204–212.
150. Wagstaff MJ, Collaço-Moraes Y, Smith J, de Belleroche JS, 169. Schlesinger MJ. How the cell copes with stress and the
Coffin RS, Latchman DS. Protection of neuronal cells from function of heat shock proteins. Pediatr Res. 1994;36(1, pt
apoptosis by Hsp27 delivered with a herpes simplex virus- 1):1–6.
based vector. J Biol Chem. 1999;274:5061–5069. 170. Rosen A, Casciola-Rosen L, Ahearn J. Novel packages of viral
151. Yanase K, Smith RM, Puccetti A, Jarett L, Madaio MP. and self-antigens are generated during apoptosis. J Exp Med.
Receptor-mediated cellular entry of nuclear localizing anti- 1995;181:1557–1561.
DNA antibodies via myosin 1. J Clin Invest. 1997;100:25–31. 171. Klareskog L, Rönnelid J, Lundberg K, Padyukov L, Alfredsson
152. Sisto M, Lisi S, D’Amore S, D’Amore M. Autoantibodies, L. Immunity to citrullinated proteins in rheumatoid arthritis.
human Fcc receptors, and autoimmunity. J Receptor Ligand Annu Rev Immunol. 2008;26:651–675.
Channel Res. 2009;2:45–57.
172. Sokolove J, Lindstrom TM, Robinson WH. Development and
153. Tezel G, Seigel GM, Wax MB. Autoantibodies to small heat deployment of antigen arrays for investigation of B-cell fine
shock proteins in glaucoma. Invest Ophthalmol Vis Sci. specificity in autoimmune disease. Front Biosci (Elite Ed).
1998;39:2277–2287. 2012;4:320–330.
154. Wax MB, Yang J, Tezel G, Peng G, Patil RV, Calkins DJ. A 173. Arbuckle MR, Schilling AR, Harley JB, James JA. A limited
model of experimental autoimmune glaucoma in rats elicited lupus anti-spliceosomal response targets a cross-reactive,
by immunization with heat shock protein27 [E-abstract proline-rich motif. J Autoimmun. 1998;11:431–438.
2884]. Invest Ophthalmol Vis Sci. 2002;43.
174. Romano C, Li Z, Arendt A, Hargrave PA, Wax MB. Epitope
155. Cauwe B, Martens E, Proost P, Opdenakker G. Multidimen- mapping of anti-rhodopsin antibodies from patients with
sional degradomics identifies systemic autoantigens and normal pressure glaucoma. Invest Ophthalmol Vis Sci. 1999;
intracellular matrix proteins as novel gelatinase B/MMP-9 40:1275–1280.
substrates. Integr Biol. 2009;1:404–426.
175. Kampylafka EI, Routsias JG, Alexopoulos H, Dalakas MC,
156. Ram M, Sherer Y, Shoenfeld Y. Matrix metalloproteinase-9 and Moutsopoulos HM, Tzioufas AG. Fine specificity of antibodies
autoimmune diseases. J Clin Immunol. 2006;26:299–307. against AQP4: epitope mapping reveals intracellular epi-
157. Yuan L, Neufeld AH. Activated microglia in the human topes. J Autoimmun. 2011;36:221–227.
glaucomatous optic nerve head. J Neurosci Res. 2001;64: 176. Vyshkina T, Kalman B. Autoantibodies and neurodegenera-
523–532. tion in multiple sclerosis. Lab Invest. 2008;88:796–807.
158. Yan X, Tezel G, Wax MB, Edward DP. Matrix metalloprotei- 177. Wright B, Warrington AE, Edberg DE, Rodriguez M. Cellular
nases and tumor necrosis factor alpha in glaucomatous optic mechanisms of central nervous system repair by natural
nerve head. Arch Ophthalmol. 2000;118:666–673. autoreactive monoclonal antibodies. Arch Neurol. 2009;66:
159. Meyerhof O, Lohmann K. Über die enzymatische Gleichge- 1456–1459.
wichtsreaktion zwischen Hexosediphosphorsäure und Diox- 178. Shoenfeld Y, Toubi E. Protective autoantibodies: role in
yacetonphosphorsäure. Naturwissenschaften. 1934;22:220. homeostasis, clinical importance, and therapeutic potential.
160. Romano C, Barrett DA, Li Z, Pestronk A, Wax MB. Anti- Arthritis Rheum. 2005;52:2599–2606.
rhodopsin antibodies in sera from patients with normal- 179. Grus F, Sun D. Immunological mechanisms in glaucoma.
pressure glaucoma. Invest Ophthalmol Vis Sci. 1995;36: Semin Immunopathol. 2008;30:121–126.
1968–1675.
180. Poletaev AB, Stepanyuk VL, Gershwin ME. Integrating
161. Joachim SC, Pfeiffer N, Grus FH. Autoantibodies in patients immunity: the immunculus and self-reactivity. J Autoimmun.
with glaucoma: a comparison of IgG serum antibodies against 2008;30:68–73.
retinal, optic nerve, and optic nerve head antigens. Graefes
Arch Clin Exp Ophthalmol. 2005;243:817–823. 181. Wax MB, Tezel G. Immunoregulation of retinal ganglion cell
fate in glaucoma. Exp Eye Res. 2009;88:825–830.
162. Grus FH, Joachim SC, Hoffmann EM, Pfeiffer N. Complex
autoantibody repertoires in patients with glaucoma. Mol Vis. 182. Grus FH, Joachim SC, Wuenschig D, Rieck J, Pfeiffer N.
2004;10:132–137. Autoimmunity and glaucoma. J Glaucoma. 2008;17:79–84.
163. Grus FH, Joachim SC, Bruns K, Lackner KJ, Pfeiffer N, Wax 183. Newman E, Reichenbach A. The Müller cell: a functional
MB. Serum autoantibodies to alpha-fodrin are present in element of the retina. Trends Neurosci. 1996;19:307–312.
glaucoma patients from Germany and the United States. 184. Newman EA, Zahs KR. Modulation of neuronal activity by
Invest Ophthalmol Vis Sci. 2006;47:968–976. glial cells in the retina. J Neurosci. 1998;18:4022–4028.
164. Tezel G, Edward DP, Wax MB. Serum autoantibodies to optic 185. Ramı́rez JM, Triviño A, Ramı́rez AI, Salazar JJ, Garcı́a-Sánchez
nerve head glycosaminoglycans in patients with glaucoma. J. Structural specializations of human retinal glial cells. Vis
Arch Ophthalmol. 1999;917–924. Res. 1996;36:2029–2036.

Downloaded from iovs.arvojournals.org on 02/05/2019


2406 Rieck IOVS, March 2013, Vol. 54, No. 3

186. Tsacopoulos M, Poitry-Yamate CL, Poitry S, Perrotet P, 208. Weiner H. A shift from adaptive to innate immunity: a
Veuthey AL. The nutritive function of glia is regulated by potential mechanism of disease progression in multiple
signals released by neurons. Glia. 1997;21:84–91. sclerosis. J Neurol. 2008;255:3–11.
187. Wang X, Tay SS, Ng YK. An immunohistochemical study of 209. Tezel G. TNF-alpha signaling in glaucomatous neurodegener-
neuronal and glial cell reactions in retinae of rats with ation. Prog Brain Res. 2008;173:409–421.
experimental glaucoma. Exp Brain Res. 2000;132:476– 210. Yuan L, Neufeld AH. Tumor necrosis factor-a: a potentially
484. neurodestructive cytokine produced by glia in the human
188. Napoli I, Neumann H. Microglial clearance function in health glaucomatous optic nerve head. Glia. 2000;32:42–50.
and disease. Neuroscience. 2009;158:1030–1038. 211. Bai Y, Shi Z, Zhuo Y, et al. In glaucoma the upregulated
189. Hung J, Chansard M, Ousman SS, Nguyen MD, Colicos MA. truncated TrkC.T1 receptor isoform in glia causes increased
Activation of microglia by neuronal activity: results from a TNF-alpha production, leading to retinal ganglion cell death.
new in vitro paradigm based on neuronal-silicon interfacing Invest Ophthalmol Vis Sci. 2010;51:6639–6651.
technology. Brain Behav Immun. 2010;24:31–40. 212. Fan BJ, Liu K, Wang DY, et al. Association of polymorphisms
190. Davalos D, Grutzendler J, Yang G, et al. ATP mediates rapid of tumor necrosis factor and tumor protein p53 with primary
microglial response to local brain injury in vivo. Nat open-angle glaucoma. Invest Ophthalmol Vis Sci. 2010;51:
Neurosci. 2005;8:752–758. 4110–4116.
191. Haynes SE, Hollopeter G, Yang G, et al. The P2Y12 receptor 213. Khan MI, Micheal S, Rana N, et al. Association of tumor
regulates microglial activation by extracellular nucleotides. necrosis factor alpha gene polymorphism G-308A with
Nat Neurosci. 2006;9:1512–1519. pseudoexfoliative glaucoma in the Pakistani population.
192. Ladeby R, Wirenfeldt M, Dalmau I, et al. Proliferating resident Mol Vis. 2009;15:2861–2867.
microglia express the stem cell antigen CD34 in response to 214. Razeghinejad MR, Rahat F, Kamali-Sarvestani E. Association of
acute neural injury. Glia. 2005;50:121–131. TNFA 308 G/A and TNFRI þ36 A/G gene polymorphisms
193. Meyer-Luehmann M, Spires-Jones TL, Prada C, et al. Rapid with glaucoma. Ophthalmic Res. 2009;42:118–124.
appearance and local toxicity of amyloid-beta plaques in a 215. Mossböck G, Renner W, El-Shabrawi Y, et al. TNF-alpha 308
mouse model of Alzheimer’s disease. Nature. 2008;451:720– G>A and 238 G>A polymorphisms are not major risk
724. factors in Caucasian patients with exfoliation glaucoma. Mol
194. Inman DM, Horner PJ. Reactive nonproliferative gliosis Vis. 2009;15:518–522.
predominates in a chronic mouse model of glaucoma. Glia. 216. Tekeli O, Turacli ME, Egin Y, Akar N, Elhan AH. Tumor
2007;55:942–953. necrosis factor alpha-308 gene polymorphism and pseudoex-
foliation glaucoma. Mol Vis. 2008;14:1815–1818.
195. Prasanna G, Krishnamoorthy R, Yorio T. Endothelin, astro-
cytes and glaucoma. Exp Eye Res. 2011;93:170–177. 217. Mattson MP, Pederson WA, Duan W, Culmsee C, Camandola S.
Cellular and molecular mechanisms underlying perturbed
196. Rappert A, Bechmann I, Pivneva T, et al. CXCR3-dependent
energy metabolism and neuronal degeneration in Alzheimer’s
microglial recruitment is essential for dendrite loss after brain
and Parkinson’s diseases. Ann N Y Acad Sci. 1999;893:154–
lesion. J Neurosci. 2004;24:8500–8509.
175.
197. Cardona AE, Pioro EP, Sasse ME, et al. Control of microglial
218. Lin MT, Beal MF. Mitochondrial dysfunction and oxidative
neurotoxicity by the fractalkine receptor. Nat Neurosci.
stress in neurodegenerative diseases. Nature. 2006;443:787–
2006;9:917–924.
795.
198. Biber K, Neumann H, Inoue K, Boddeke HW. Neuronal On
219. Musunuru K, Darnell RB. Paraneoplastic neurologic disease
and Off signals control microglia. Trends Neurosci. 2007;30:
antigens: RNA-binding proteins and signaling proteins in
596–602.
neuronal degeneration. Annu Rev Neurosci. 2001;24:239–
199. Tezel GMD, Wax MB. Glial modulation of retinal ganglion dell 262.
death in glaucoma. J Glaucoma. 2003;12:63–68. 220. Solimena M, De Camilli P. Synaptic autoimmunity and the
200. Tezel G, Chauhan BC, LeBlanc RP, Wax MB. Immunohisto- Salk factor. Neuron. 2000;28:309–316.
chemical assessment of the glial mitogen-activated protein 221. Maruyama I, Nakazawa M, Ohguro H. Autoimmune mecha-
kinase activation in glaucoma. Invest Ophthalmol Vis Sci. nisms in molecular pathology of glaucomatous optic
2003;44:3025–3033. neuropathy [in Japanese]. Nippon Ganka Gakkai Zasshi.
201. Pasinetti GM. Cyclooxygenase and inflammation in Alz- 2001;105:205–212.
heimer’s disease: experimental approaches and clinical 222. Schori H, Yoles E, Wheeler LA, Raveh T, Kimchi A, Schwartz
interventions. J Neurosci Res. 1998;54:1–6. M. Immune-related mechanisms participating in resistance
202. Gonzalez-Scarano F, Baltuch G. Microglia as mediators of and susceptibility to glutamate toxicity. Eur J Neurosci. 2002;
inflammatory and degenerative diseases. Annu Rev Neurosci. 16:557–564.
1999;22:219–240. 223. Wax MB, Yang J, Tezel G. Serum autoantibodies in patients
203. Julien J-P. Amyotrophic lateral sclerosis: unfolding the toxicity with glaucoma. J Glaucoma. 2001;10(5)(suppl 1):S22–S24.
of the misfolded. Cell. 2001;104:581–591. 224. Albert M, Austin L, Darnell RB. Detection and treatment of
204. Venters HD, Tang Q, Liu Q, VanHoy RW, Dantzer R, Kelley activated T cells in the cerebrospinal fluid of patients with
KW. A new mechanism of neurodegeneration: a proinflam- paraneoplastic cerebellar degeneration. Ann Neurol. 2000;
matory cytokine inhibits receptor signaling by a survival 47:9–17.
peptide. Proc Natl Acad Sci U S A. 1999;96:9879–9884. 225. Fearon DT, Locksley RM. The instructive role of innate
205. Friedlander RM. Role of caspase 1 in neurologic disease. Arch immunity in the acquired immune response. Science. 1996;
Neurol. 2000;57:1273–1276. 272:50–53.
206. González-Scarano F, Baltuch G. Microglia as mediators of 226. Li Y, Ucccelli A, Laxer KD, et al. Local-clonal expansion of
inflammatory and degenerative diseases. Annu Rev Neurosci. infiltrating T lymphocytes in chronic encephalitis of Rasmus-
1999;22:219–240. sen. J Immunol. 1997;158:1428–1437.
207. Hickey WF, Kimura H. Perivascular microglial cells of the 227. Neumann H, Medana IM, Bauer J, Lassmann H. Cytotoxic T
CNS are bone marrow-derived and present antigen in vivo. lymphocytes in autoimmune and degenerative CNS diseases.
Science. 1988;239:290–292. Trends Neurosci. 2002;25:313–319.

Downloaded from iovs.arvojournals.org on 02/05/2019


IOVS, March 2013, Vol. 54, No. 3 A Summary of the Current Knowledge 2407

228. Caspi R. Autoimmunity in the immune privileged eye: 251. Hajishengallis G, Lambris JD. Crosstalk pathways between
pathogenic and regulatory T cells. Immunol Res. 2008;42: Toll-like receptors and the complement system. Trends
41–50. Immunol. 2010;31:154–163.
229. Nielsen HH, Ladeby R, Fenger C, et al. Enhanced microglial 252. Beutler B. Microbe sensing, positive feedback loops, and the
clearance of myelin debris in T cell-infiltrated central nervous pathogenesis of inflammatory diseases. Immunol Rev. 2009;
system. J Neuropathol Exp Neurol. 2009;68:845–856. 227:248–263.
230. Streilein JW. Unraveling immune privilege. Science. 1995; 253. Butchi N, Du MB, Peterson KE. Interactions between TLR7
270:1158–1159. and TLR9 agonists and receptors regulate innate immune
231. Albert ML, Darnell JC, Bender A, Francisco LM, Bhardwaj N, responses by astrocytes and microglia. Glia. 2009;58:650–
Darnell RB. Tumor-specific killer cells in paraneoplastic 664.
cerebellar degeneration. Nat Med. 1998;4:1321–1324. 254. Heil F, Hemmi H, Hochrein H, et al. Species-specific
232. Whitney KD, Andrews PI, McNamara JO. Immunoglobulin G recognition of single-stranded RNA via Toll-like receptors 7
and complement immunoreactivity in the cerebral cortex of and 8. Science. 2004;303:1526–1529.
patients with Rasmussen’s encephalitis. Neurology. 1999;53: 255. Luo C, Yang X, Powell DW, Klein JB, Tezel G. Stress proteins
699–708. and immunostimulatory signaling through toll-like receptors
233. Abbott N. Astrocyte-endothelial interactions and blood-brain in glaucoma [E-abstract 4048]. Invest Ophthalmol Vis Sci.
barrier permeability. J Anat. 2002;200:527. 2009;50.
234. Grieshaber MC, Flammer J. Does the blood-brain barrier play 256. Tsan MF, Gao B. Heat shock proteins and immune system. J
a role in glaucoma? Surv Ophthalmol. 2007;52(6)(suppl 1): Leukoc Biol. 2009;85:905–910.
S115–S121. 257. Wallin RP, Lundqvist A, Moré SH, von Bonin A, Kiessling R,
235. Goverman J. Autoimmune T cell responses in the central Ljunggren HG. Heat-shock proteins as activators of the innate
nervous system. Nat Rev Immunol. 2009;9:393–407. immune system. Trends Immunol. 2002;23:130–135.
236. Nguyen MD, Julien JP, Rivest S. Innate immunity: the missing 258. Roelofs MF, Boelens WC, Joosten LA, et al. Identification of
link in neuroprotection and neurodegeneration? Nat Rev small heat shock protein B8 (HSP22) as a novel TLR4 ligand
Neurosci. 2002;3:216–227. and potential involvement in the pathogenesis of rheumatoid
arthritis. J Immunol. 2006;176:7021–7027.
237. Ransohoff RM, Kivisakk P, Kidd G. Three or more routes for
259. Tükel Ç. CsgA is a pathogen-associated molecular pattern of
leukocyte migration into the central nervous system. Nat Rev
Salmonella enterica serotype Typhimurium that is recognized
Immunol. 2003;3:569–581.
by Toll-like receptor 2. Mol Microbiol. 2005;58:289–304.
238. Huber JD, Egleton RD, Davis TP. Molecular physiology and
260. Ohashi K, Burkart V, Flohé S, Kolb H. Cutting edge: heat
pathophysiology of tight junctions in the blood-brain barrier.
shock protein 60 is a putative endogenous ligand of the Toll-
Trends Neurosci. 2001;24:719–725.
like receptor-4 complex. J Immunol. 2000;164:558–561.
239. Howell GR, Soto I, Zhu X, et al. Radiation treatment inhibits
261. Johnson JD, Fleshner M. Releasing signals, secretory path-
monocyte entry into the optic nerve head and prevents
ways, and immune function of endogenous extracellular heat
neuronal damage in a mouse model of glaucoma. J Clin
shock protein 72. J Leukoc Biol. 2006;79:425–434.
Invest. 2012;122:1246–1261.
262. Young DB. Heat-shock proteins: immunity and autoimmunity.
240. Xu H, Dawson R, Forrester JV, Liversidge J. Identification of
Curr Opin Immunol. 1992;4:396–400.
novel dendritic cell populations in normal mouse retina.
Invest Ophthalmol Vis Sci. 2007;48:1701–1710. 263. Young RA, Elliott TJ. Stress proteins, infection, and immune
surveillance. Cell. 1989;59:5–8.
241. Colonna M. TREMs in the immune system and beyond. Nat
Rev Immunol. 2003;3:445–453. 264. Kol A, Lichtman AH, Finberg RW, Libby P, Kurt-Jones EA.
Cutting edge: heat shock protein (HSP) 60 activates the
242. Olson JK, Miller SD. Microglia initiate central nervous system innate immune response: CD14 is an essential receptor for
innate and adaptive immune responses through multiple HSP60 activation of mononuclear cells. J Immunol. 2000;
TLRs. J Immunol. 2004;173:3916–3924. 164:13–17.
243. Lehnardt S. Innate immunity and neuroinflammation in the 265. Liu D, Yumoto D, Hirota K, et al. Histone-like DNA binding
CNS: the role of microglia in Toll-like receptor-mediated protein of Streptococcus intermedius induces the expression
neuronal injury. Glia. 2009;58:253–263. of pro-inflammatory cytokines in human monocytes via
244. Akira S, Yamamoto M, Takeda K. Role of adapters in Toll-like activation of ERK1/2 and JNK pathways. Cell Microbiol.
receptor signalling. Biochem Soc Trans. 2003;31(pt 3):637– 2008;10:262–276.
642. 266. Rajaiah R, Moudgil KD. Heat-shock proteins can promote as
245. Ford JW, McVicar DW. TREM and TREM-like receptors in well as regulate autoimmunity. Autoimmun Rev. 2009;8:
inflammation and disease. Curr Opin Immunol. 2009;21:38– 388–393.
46. 267. Damian RT. Molecular mimicry in biological adaptation.
246. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflamma- Science. 1965;147:824.
tory responses can be triggered by TREM-1, a novel receptor 268. Damian RT. Molecular mimicry: antigen sharing by parasite
expressed on neutrophils and monocytes. J Immunol. 2000; and host and its consequences. Am Naturalist. 1964;98:129–
164:4991–4995. 149.
247. Sharif O, Knapp S. From expression to signaling: roles of 269. Galloway PH, Warner SJ, Morshed MG, Mikelberg FS.
TREM-1 and TREM-2 in innate immunity and bacterial Helicobacter pylori infection and the risk for open-angle
infection. Immunobiology. 2008;213:701–713. glaucoma. Ophthalmology. 2003;110:922–925.
248. Ito H, Hamerman JA. TREM-2, triggering receptor expressed 270. Kountouras J, Zavos C, Chatzopoulos D. Induction of
on myeloid cell-2, negatively regulates TLR responses in apoptosis as a proposed pathophysiological link between
dendritic cells. Eur J Immunol. 2012;42:176–185. glaucoma and Helicobacter pylori infection. Med Hypoth.
249. Janeway CA, Medzhitov R. Innate immune recognition. Annu 2004;62:378–381.
Rev Immunol. 2002;20:197–216. 271. Kountouras J, Mylopoulos N, Konstas AG, Zavos C, Chatzo-
250. Choe J, Kelker MS, Wilson IA. Crystal structure of human Toll- poulos D, Boukla A. Increased levels of Helicobacter pylori
like receptor 3 (TLR3) ectodomain. Science. 2005;309:581–585. IgG antibodies in aqueous humor of patients with primary

Downloaded from iovs.arvojournals.org on 02/05/2019


2408 Rieck IOVS, March 2013, Vol. 54, No. 3

open-angle and exfoliation glaucoma. Graefes Arch Clin Exp 293. Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a
Ophthalmol. 2003;241:884–890. key system for immune surveillance and homeostasis. Nat
272. Cohen IR. Antigenic mimicry, clonal selection and autoim- Immunol. 2010;11:785–797.
munity. J Autoimmun. 2001;16:337–340. 294. Kulkarni AP, Kellaway LA, Lahiri DK, Kotwal GL. Neuropro-
273. Oldstone MB. Molecular mimicry, microbial infection, and tection from complement-mediated inflammatory damage.
autoimmune disease: evolution of the concept. Curr Top Ann N Y Acad Sci. 2004;1035:147–164.
Microbiol Immunol. 2005;296:1–17. 295. Neufeld AH, Liu B. Glaucomatous optic neuropathy: when
274. Kim JM, Kim SH, Park KH, Han SY, Shim HS. Investigation of glia misbehave. Neuroscientist. 2003;9:485–495.
the association between Helicobacter pylori infection and 296. Tezel G, Wax MB. Increased production of tumor necrosis
normal tension glaucoma. Invest Ophthalmol Vis Sci. 2011; factor-alpha by glial cells exposed to simulated ischemia or
52:665–668. elevated hydrostatic pressure induces apoptosis in cocul-
275. Rodriguez D, Morrison CJ, Overall CM. Matrix metalloprotei- tured retinal ganglion cells. J Neurosci. 2000;20:8693–8700.
nases: what do they not do? New substrates and biological 297. Tezel G, Wax MB. Hypoxia-inducible factor 1alpha in the
roles identified by murine models and proteomics. Biochim glaucomatous retina and optic nerve head. Arch Ophthal-
Biophys Acta. 2010;1803:39–54. mol. 2004;122:1348–1356.
276. Kurien BT, Scofield RH. Autoimmunity and oxidatively 298. Boulanger LM. Immune proteins in brain development and
modified autoantigens. Autoimmun Rev. 2008;7:567–573. synaptic plasticity. Neuron. 2009;64:93–109.
277. Chiba S, Yokota S, Yonekura K, et al. Autoantibodies against 299. Rosen AM, Stevens B. The role of the classical complement
HSP70 family proteins were detected in the cerebrospinal cascade in synapse loss during development and glaucoma.
fluid from patients with multiple sclerosis. J Neurol Sci. In: Cohen IR, Lajtha A, Lambris JD, Paoletti R, eds.
2006;241:39–43. Inflammation and Retinal Disease: Complement Biology
278. Yonekura K, Yokota S, Tanaka S, et al. Prevalence of anti-heat and Pathology. Vol. 73. Berlin: Springer; 2010:75–93.
shock protein antibodies in cerebrospinal fluids of patients 300. Katz LC, Shatz CJ. Synaptic activity and the construction of
with Guillain-Barre syndrome. J Neuroimmunol. 2004;156: cortical circuits. Science. 1996;274:1133–1138.
204–209. 301. Singhrao SK, Neal JW, Rushmere NK, Morgan BP, Gasque P.
279. Gasque P. Complement: a unique innate immune sensor for Spontaneous classical pathway activation and deficiency of
danger signals. Mol Immunol. 2004;41:1089–1098. membrane regulators render human neurons susceptible to
280. Howell GR. Molecular clustering identifies complement and complement lysis. Am J Pathol. 2000;157:905–918.
endothelin induction as early events in a mouse model of 302. Huh GS, Boulanger LM, Du H, Riquelme PA, Brotz TM, Shatz
glaucoma. J Clin Invest. 2011;121:1429–1444. CJ. Functional requirement for class I MHC in CNS
281. Stevens B, Allen NJ, Vazquez LE, et al. The classical development and plasticity. Science. 2000;290:2155–2159.
complement cascade mediates CNS synapse elimination. 303. Neumann H, Cavalié A, Jenne DE, Wekerle H. Induction of
Cell. 2007;131:1164–1178. MHC class I genes in neurons. Science. 1995;269:549–552.
282. Ikeda Y, Maruyama I, Nakazawa M, Ohguro H. Clinical 304. Stasi K, Nagel D, Yang X, et al. Complement component 1Q
significance of serum antibody against neuron-specific (C1Q) upregulation in retina of murine, primate, and human
enolase in glaucoma patients. Jpn J Ophthalmol. 2002;46: glaucomatous eyes. Invest Ophthalmol Vis Sci. 2006;47:
13–17. 1024–1029.
283. Buckley C, Vincent A. Autoimmune channelopathies. Nat 305. Howell GR, Libby RT, Jakobs TC, et al. Axons of retinal
Clin Pract Neurol. 2005;1:22–33. ganglion cells are insulted in the optic nerve early in DBA/2J
284. Scofield RH. Autoantibodies as predictors of disease. Lancet. glaucoma. J Cell Biol. 2007;179:1523–1537.
2004;363:1544–1546. 306. Sigal IA, Ethier CR. Biomechanics of the optic nerve head.
285. Arbuckle MR, McClain MT, Rubertone MV, et al. Develop- Exp Eye Res. 2009;88:799–807.
ment of autoantibodies before the clinical onset of systemic 307. Ambrosini E, Aloisi F. Chemokines and glial cells: a complex
lupus erythematosus. N Engl J Med. 2003;349:1526–1533. network in the central nervous system. Neurochem Res.
286. Quintana FJ, Cohen IR. The natural autoantibody repertoire 2004;29:1017–1038.
and autoimmune disease. Biomed Pharmacother. 2004;58: 308. Langmann T. Microglia activation in retinal degeneration. J
276–281. Leukoc Biol. 2007;81:1345–1351.
287. Lennon VA, Wingerchuk DM, Kryzer TJ, et al. A serum 309. Nakazawa T, Nakazawa C, Matsubara A, et al. Tumor necrosis
autoantibody marker of neuromyelitis optica: distinction factor-a mediates oligodendrocyte death and delayed retinal
from multiple sclerosis. Lancet. 2004;364:2106–2112. ganglion cell loss in a mouse model of glaucoma. J Neurosci.
288. Dziewas R, Kis B, Grus FH, Zimmermann CW. Antibody 2006;26:12633–12641.
pattern analysis in the Guillain-Barré syndrome and patho- 310. Hawkins BT, Davis TP. The blood-brain barrier/neurovascular
logic controls. J Neuroimmunol. 2001;119:287–296. unit in health and disease. Pharmacol Rev. 2005;57:173–185.
289. Baroni SS, Santillo M, Bevilacqua F, et al. Stimulatory 311. Yong VW, Marks S. The interplay between the immune and
autoantibodies to the PDGF receptor in systemic sclerosis. central nervous systems in neuronal injury. Neurology. 2010;
N Engl J Med. 2006;354:2667–2676. 74(suppl 1);S9–S16.
290. Kubo T, Uchida Y, Watanabe Y, et al. Augmented TLR9- 312. Ahmed F, Brown KM, Stephen DA, Morrison JC, Johnson EC,
induced Btk activation in PIR-B-deficient B-1 cells provokes Tomarev SI. Microarray analysis of changes in mRNA levels in
excessive autoantibody production and autoimmunity. J Exp the rat retina after experimental elevation of intraocular
Med. 2009;206:1971–1982. pressure. Invest Ophthalmol Vis Sci. 2004;45:1247–1258.
291. Hueber W, Tomooka BH, Batliwalla F, et al. Blood autoanti- 313. Taylor PR, Carugati A, Fadok VA. A hierarchical role for
body and cytokine profiles predict response to anti-tumor classical pathway complement proteins in the clearance of
necrosis factor therapy in rheumatoid arthritis. Arthritis Res apoptotic cells in vivo. J Exp Med. 2000;192:359–366.
Ther. 2009;11:R76. 314. Botto M, Dell’Agnola C, Bygrave AE, et al. Homozygous C1q
292. Walport MJ. Complement. N Engl J Med. 2001;344:1058– deficiency causes glomerulonephritis associated with multi-
1066. ple apoptotic bodies. Nat Genet. 1998;19:56–59.

Downloaded from iovs.arvojournals.org on 02/05/2019


IOVS, March 2013, Vol. 54, No. 3 A Summary of the Current Knowledge 2409

315. Cowell RM, Plane JM, Silverstein FS. Complement activation 333. Ghaffariyeh A, Honarpisheh N, Heidari MH, Puyan S, Abasov
contributes to hypoxic-ischemic brain injury in neonatal rats. F. Brain-derived neurotrophic factor as a biomarker in primary
J Neurosci. 2003;23:9459–9468. open-angle glaucoma. Optom Vis Sci. 2011;88:80–85.
316. Rancan M, Morganti-Kossman MC, Barnum SR, et al. Central 334. Ferreira SM, Lerner SF, Brunzini R, Evelson PA, Llesuy SF.
nervous system-targeted complement inhibition mediates Oxidative stress markers in aqueous humor of glaucoma
neuroprotection after closed head injury in transgenic mice. J patients. Am J Ophthalmol. 2004;137:62–69.
Cereb Blood Flow Metab. 2003;23:1070–1074. 335. Erdurmus M, Yağcı R, Atis Ö, Karadağ R, Akbas A, Hepsen IF.
317. Yamada K, Miwa T, Liu J, Nangaku M, Song WC. Critical Antioxidant status and oxidative stress in primary open angle
protection from renal ischemia reperfusion injury by CD55 glaucoma and pseudoexfoliative glaucoma. Curr Eye Res.
and CD59. J Immunol. 2004;172:3869–3875. 2011;36:713–718.
318. Schäfer MK, Schwaeble WJ, Post C, et al. Complement C1q is 336. Tezel G. Oxidative stress in glaucomatous neurodegenera-
dramatically up-regulated in brain microglia in response to tion: mechanisms and consequences. Prog Retin Eye Res.
transient global cerebral ischemia. J Immunol. 2000;164: 2006;25:490–513.
5446–5452. 337. Moreno MC, Campanelli J, Sande P, Sánez DA, Keller
319. Dangond F, Hwang D, Camelo S, et al. Molecular signature of Sarmiento MI, Rosenstein RE. Retinal oxidative stress
late-stage human ALS revealed by expression profiling of induced by high intraocular pressure. Free Radic Biol
postmortem spinal cord gray matter. Physiol Genomics. Med. 2004;37:803–812.
2004;16:229–239. 338. Hinerfeld D, Traini MD, Weinberger RP, et al. Endogenous
320. Fonseca MI, Kawas CH, Troncoso JC, Tenner AJ. Neuronal mitochondrial oxidative stress: neurodegeneration, proteo-
localization of C1q in preclinical Alzheimer’s disease. Neuro- mic analysis, specific respiratory chain defects, and effica-
biol Dis. 2004;15:40–46. cious antioxidant therapy in superoxide dismutase 2 null
mice. J Neurochem. 2004;88:657–667.
321. Stasi K, Nagel D, Yang X, et al. Complement component 1Q
(C1Q) upregulation in retina of murine, primate, and human 339. Miyara N, Shinzato M, Yamashiro Y, Iwamatsu A, Kariya K,
glaucomatous eyes. Invest Ophthalmol Vis Sci. 2006;47: Sawaguchi S. Proteomic analysis of rat retina in a steroid-
1024–1029. induced ocular hypertension model: potential vulnerability
to oxidative stress. Jpn J Ophthalmol. 2008;52:84–90.
322. Johnson EC, Jia L, Cepurna WO, Doser TA, Morrison JC.
Global changes in optic nerve head gene expression after 340. Wilson JX. Antioxidant defense of the brain: a role for
exposure to elevated intraocular pressure in a rat glaucoma astrocytes. Can J Physiol Pharmacol. 1997;75:1149–1163.
model. Invest Ophthalmol Vis Sci. 2007;48:3161–3177. 341. Engin KN, Engin G, Kucuksahin H, Oncu M, Engin G,
323. Miyahara T, Kikuchi T, Akimoto M, Kurokawa T, Shibuki H, Guvener B. Clinical evaluation of the neuroprotective effect
Yoshimura N. Gene microarray analysis of experimental of alpha-tocopherol against glaucomatous damage. Eur J
glaucomatous retina from cynomologus monkey. Invest Ophthalmol. 2007;17:528–533.
Ophthalmol Vis Sci. 2003;44:4347–4356. 342. Neufeld AH, Hernandez MR, Gonzalez M. Nitric oxide
324. Zhang X, Kimura Y, Fang C, et al. Regulation of Toll-like synthase in the human glaucomatous optic nerve head. Arch
Ophthalmol. 1997;115:497–503.
receptor-mediated inflammatory response by complement in
vivo. Blood. 2007;110:228–236. 343. Ritch R. Neuroprotection: is it already applicable to glaucoma
therapy? Curr Opin Ophthalmol. 2000;11:78–84.
325. Kim DD, Song W-C. Membrane complement regulatory
proteins. Clin Immunol. 2006;118:127–136. 344. Liu B, Neufeld AH. Nitric oxide synthase-2 in human optic
nerve head astrocytes induced by elevated pressure in vitro.
326. Wax MB. The case for autoimmunity in glaucoma. Exp Eye
Arch Ophthalmol. 2001;119:240–245.
Res. 2011;93:187–190.
345. Liu B, Neufeld, Arthur H. Expression of nitric oxide synthase-
327. Flavell RA, Hafler DA. Autoimmunity: what is the turning
2 (NOS-2) in reactive astrocytes of the human glaucomatous
point?: editorial overview. Curr Opin Immunol. 1999;11:
optic nerve head. Glia. 2000;30:178–186.
635–637.
346. Mozaffarieh M, Grieshaber MC, Flammer J. Oxygen and blood
328. Hafler DA, Flavell R. Autoimmunity. How to know thy self. flow: players in the pathogenesis of glaucoma. Mol Vis. 2008;
Curr Opin Immunol. 1996;8:805–807. 14:224–233.
329. Roh M, Zhang Y, Murakami Y, et al. Etanercept, a widely used 347. Sacca S, Pascotto A, Camicione P, Capris P, Izzotti A.
inhibitor of tumor necrosis factor-alpha (TNF-alpha), prevents Oxidative DNA damage in the human trabecular meshwork:
retinal ganglion cell loss in a rat model of glaucoma. PLoS One. clinical correlation in patients with primary open-angle
2012;7:e40065. glaucoma. Arch Ophthalmol. 2005;123:458–463.
330. Ognibene A, Ciuti R, Tozzi P, Messeri G. Maternal serum 348. McElnea EM, Quill B, Docherty NG, et al. Oxidative stress,
superoxide dismutase (SOD): a possible marker for screening mitochondrial dysfunction and calcium overload in human
Down syndrome affected pregnancies. Prenat Diagn. 1999; lamina cribrosa cells from glaucoma donors. Mol Vis. 2011;
19:1058–1060. 17:1182–1191.
331. Golubnitschaja O, Flammer J. What are the biomarkers for 349. Pinazo-Durán MD, Zanón-Moreno V, Garcı́a-Medina JJ, Galle-
glaucoma? Surv Ophthalmol. 2007;52(suppl 2):S155–S161. go-Pinazo R. Evaluation of presumptive biomarkers of
332. Grus FH, Joachim SC, Sandmann S, et al. Transthyretin and oxidative stress, immune response and apoptosis in primary
complex protein pattern in aqueous humor of patients with open-angle glaucoma. Curr Opin Pharmacol. 2012;13:98–
primary open-angle glaucoma. Mol Vis. 2008;14:1437–1445. 107.

Downloaded from iovs.arvojournals.org on 02/05/2019

You might also like