Manuscript_Van de Velde et al PRONEU-D-14-00098

advertisement
1
Towards axonal regeneration and neuroprotection in glaucoma: Rho kinase inhibitors as promising
2
therapeutics
3
Sarah Van de Velde1, Lies De Groef², Ingeborg Stalmans1, Lieve Moons2 and Inge Van Hove2
4
5
1
6
2
7
Section, Department of Biology, KU Leuven, Leuven, Belgium
Laboratory of Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium
Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology
8
9
Corresponding author:
10
Dr. Lieve Moons
11
Neural Circuit Development and Regeneration Research Group
12
Animal Physiology and Neurobiology Section
13
Department of Biology
14
KU Leuven
15
Naamsestraat 61, Box 2464
16
B-3000 Leuven, Belgium
17
Tel: +32-16-32.39.91
18
Fax: +32-16-32.42.62
19
Lieve.moons@bio.kuleuven.be
20
21
Running title: ROCK inhibition for retinal axonal regeneration and neuroprotection
22
Key words: ROCK inhibition, glaucoma, axonal regeneration, neuroprotection and ocular blood flow
23
1
24
Abstract
25
Due to a prolonged life expectancy worldwide, the incidence of age-related
26
neurodegenerative disorders such as glaucoma is increasing. Glaucoma is the second cause of
27
blindness, resulting from a slow and progressive loss of retinal ganglion cells (RGCs) and their axons.
28
Up to now, intraocular pressure (IOP) reduction is the only treatment modality by which
29
ophthalmologists attempt to control disease progression. However, not all patients benefit from this
30
therapy, and the pathophysiology of glaucoma is not always associated with an elevated IOP. These
31
limitations, together with the multifactorial etiology of glaucoma, urge the pressing medical need for
32
novel and alternative treatment strategies. Such new therapies should focus on preventing or
33
retarding RGC death, but also on repair of injured axons, to ultimately preserve or improve structural
34
and functional connectivity. In this respect, Rho-associated coiled-coil forming protein kinase (ROCK)
35
inhibitors hold a promising potential to become very prominent drugs for future glaucoma
36
treatment. Their field of action in the eye does not seem to be restricted to IOP reduction by
37
targeting the trabecular meshwork or improving filtration surgery outcome. Indeed, over the past
38
years, important progress has been made in elucidating their ability to improve ocular blood flow, to
39
prevent RGC death/increase RGC survival and to retard axonal degeneration or induce proper axonal
40
regeneration. Within this review, we aim to highlight the currently known capacity of ROCK inhibition
41
to promote neuroprotection and regeneration in several in vitro, ex vivo and in vivo experimental
42
glaucoma models.
43
44
Contents
45
I. Introduction .......................................................................................................................................... 3
46
II. Glaucoma pathophysiology ................................................................................................................. 4
47
III. Novel strategies for glaucoma therapy: neuroprotection and axonal regeneration ......................... 7
48
IV. The Rho-ROCK pathway ..................................................................................................................... 8
2
49
1. Downstream effectors of ROCK: well-known cytoskeletal regulators ............................................ 9
50
2. The ROCK signaling cascade and its involvement in neuronal cell death and axonal outgrowth . 10
51
V. ROCK inhibition as a multi-target approach for glaucoma treatment .............................................. 12
52
1. ROCK inhibitors as IOP lowering and anti-scarring agents in glaucoma ....................................... 12
53
2. ROCK inhibitors as novel neuroprotective and axon regenerative agents in glaucoma ............... 14
54
2.1. ROCK expression in the healthy and injured/diseased CNS ................................................... 15
55
2.2. ROCK inhibition as a promising tool to induce axon regeneration in the injured visual system
56
....................................................................................................................................................... 16
57
2.3. ROCK inhibition as a promising tool to support neuronal survival in the injured visual system
58
....................................................................................................................................................... 22
59
3. ROCK inhibition as promising regulators of ocular blood flow ..................................................... 26
60
V. Future perspectives: (pre-)clinical development of ROCK inhibitors for glaucoma treatment ........ 26
61
VI. Conclusion ........................................................................................................................................ 28
62
63
I. INTRODUCTION
64
The term glaucoma refers to a wide range of optic neuropathies associated with
65
degeneration of retinal ganglion cells (RGCs) and their respective axons, leading to slow progressive
66
visual field loss (Foster et al. 2002). If not adequately treated, most types of glaucoma progress
67
without obvious symptoms towards gradual loss of visual function or even blindness (Friedman et al.
68
2004). Nowadays, it is considered as the second leading cause of blindness throughout the world
69
(Resnikoff et al. 2008). This neurodegenerative disease mostly appears after the 4th decade of life,
70
and its frequency significantly increases with age. In 2010, at least 60.5 million people suffered from
71
glaucoma worldwide. This number is expected to increase up to 80 million by 2020 (Quigley and
3
72
Broman 2006). Risk factors for glaucoma not only include age, but also family history (Wolfs et al.
73
1998), gender (Rudnicka et al. 2006), ethnic background (Racette et al. 2003), severe myopia
74
(Mitchell et al. 1999), disturbed cerebrospinal fluid pressure (Fleischman and Allingham 2013),
75
vascular disorders (Bonomi et al. 2000) and importantly, an increased intraocular pressure (IOP)
76
(Boland and Quigley 2007).
77
With the currently available treatment modalities, glaucomatous visual field deterioration
78
cannot be prevented, yet lowering the IOP can slow down glaucoma disease progression
79
(VanVeldhuisen et al. 2000; Kass et al. 2002; Anastassiadis et al. 2011). Therefore glaucoma
80
treatment is mainly directed towards lowering IOP (VanVeldhuisen et al. 2000; Kass et al. 2002). A
81
sustained reduction of IOP can be achieved with topical therapy, laser therapy or surgical
82
interventions (Quigley 2011). Nevertheless, signs of progression can be seen in many patients despite
83
well-controlled IOP. Therefore, the development of neuroprotective strategies may be of high value
84
in the future treatment of this multifactorial neurodegenerative disorder. Until now, only two
85
potential neuroprotective agents have been investigated in clinical trials for glaucoma, i.e.
86
memantine and brimonidine.
87
Over the past few years, many studies have highlighted the important role of the Rho and
88
Rho-associated coiled-coil protein kinase (ROCK) pathway in the pathogenesis and treatment of
89
glaucoma. Modulation of this pathway seems to be involved in the regulation of IOP via the
90
trabecular meshwork (TM) and may also serve as a potent anti-scarring agent after glaucoma
91
surgery. More importantly, the amount of studies reporting ROCK inhibition as a very appealing
92
therapeutic approach that confers neuroprotection and axonal regeneration, substantially increased
93
over the last decade. Therefore, the purpose of this review is to summarize the current knowledge
94
on the upcoming neuroprotective and axon regenerative potential of ROCK inhibitors, which may be
95
of great importance in the development of a novel neuroprotective/regenerative strategy for
96
glaucoma therapy.
97
4
98
II. GLAUCOMA PATHOPHYSIOLOGY
99
The exact mechanisms of how glaucoma develops are still not well-known, although the
100
initiation and propagation of this disease is thought to be situated around the optic nerve head
101
(ONH). Yet, two principal hypotheses, the mechanical and ischemic/vascular theories have been
102
described (Fechtner and Weinreb 1994; Flammer et al. 2002b; Flammer and Mozaffarieh 2007)
103
(Figure 1). The classical mechanical theory suggests that the development of glaucoma is a direct
104
consequence of an increased IOP causing damage to the ONH (Yan et al. 1994). The IOP reflects the
105
balance between aqueous humor (AH) production and outflow through either the conventional
106
pathway via the TM, or the unconventional pathway via the uveoscleral route (Goel et al. 2010). IOP
107
elevation above 21 mmHg due to impaired TM function, is considered as the most important
108
measurable risk factor for the development of glaucoma (Weinreb and Khaw 2004) and believed to
109
induce damage to the RGC axons by tissue deformation at the level of the lamina cribrosa, thereby
110
leading to ONH cupping. When RGC axons exit the eye at the ONH, the lamina cribrosa provides the
111
only support and protection in an otherwise firm scleral shell, making it the most vulnerable site of
112
the retina/optic nerve to mechanical stress (Sigal and Ethier 2009). Cupping of the ONH in response
113
to elevated IOP causes compression of RGC axons, leading to a disruption of the axoplasmic transport
114
between the retina and the brain, which is essential for proper RGC survival (Flammer et al. 2002b).
115
Additionally, glial cells at the ONH shift their production of extracellular matrix (ECM) components
116
and increase their secretion of matrix metalloproteinases, thereby reducing mechanical support to
117
nerve fibers, making them even more vulnerable to elevated IOP (Hernandez 2000; Kirwan et al.
118
2004; Dahlmann-Noor et al. 2010; Quill et al. 2011; Akhter et al. 2013; De Groef et al. 2014).
119
Although generally accepted for a long time, the mechanical theory fails to explain several
120
features correlated with the appearance of glaucoma. Many patients (25-30%) suffer from
121
glaucomatous symptoms without the appearance of an increased IOP (normal tension glaucoma
122
(NTG)) and pressure reduction does not avoid glaucomatous damage in all patients with initial ocular
123
hypertension. On the other hand, some patients with ocular hypertension show no damage to the
5
124
optic nerve (Chauhan and Drance 1992; Martinez-Bello et al. 2000; Agarwal et al. 2009). These
125
observations indicate that multiple other factors, unrelated to IOP, play an important role in the
126
development of glaucoma. Increased incidence of glaucoma, especially NTG, is observed in patients
127
with systemic vascular disorders like hypotension (Hayreh et al. 1994; Hayreh 1999), cardiovascular
128
disease (Hayreh 1999), vasospastic disorders (Broadway and Drance 1998), migraine (Wang et al.
129
1997), diabetes (Mitchell et al. 1997) and cerebral ischemia (Stroman et al. 1995). Therefore, the
130
vascular theory considers a disturbed blood supply to the retina as the primary cause of optic
131
atrophy (Flammer et al. 2002b). Insufficient vascular autoregulation to adapt ocular perfusion may
132
result in a decreased retinal blood supply, causing ischemic damage (Morgan 2004). This then leads
133
to an impaired perfusion of the ONH, causing ischemic injury to the retina. Notably, the mechanical
134
and vascular theory are closely interlinked, because excavation of the ONH at the lamina cribrosa as
135
a direct consequence of elevated IOP, also leads to kinking of the retinal blood vessels (Flammer
136
1994; Flammer et al. 2002b; Flammer and Mozaffarieh 2007; Chang and Goldberg 2012b).
137
It is further hypothesized that both mechanical and/or ischemic insults to RGC axons
138
ultimately result in mitochondrial dysfunction leading to the induction of RGC apoptosis due to
139
oxidative damage (Figure 1). Mitochondrial dysfunction causes impaired energy supply, rendering
140
RGCs to exist in a lower energetic state, which results in a passive release of glutamate into the
141
extracellular space. In this state, cells are still able to transmit visual information, but are more
142
vulnerable to stress stimuli. Upon insult, also glial cells (microglia and astrocytes) will become
143
reactive, causing a gradual rise in cytokines and other factors, such as TNF-α, glutamate,
144
prostaglandins and nitric oxide. Initially, Müller glia will try to maintain a physiological balance, but
145
eventually, this process will become insufficient, resulting in an increased level of these molecules,
146
some of which, such as glutamate, are potentially toxic to neuronal cells (Toft-Kehler et al.2014).
147
Because RGCs are already in a decreased energetic state, they are more susceptible to be affected
148
and as a result, RGCs will undergo apoptosis at different rates, depending on their receptor profile
149
(Osborne et al. 2001; Osborne 2009; Chrysostomou et al. 2013). Notably, next to the mechanical and
6
150
vascular factors, inflammatory, autoimmune or even other (still unknown) factors may also
151
contribute to glaucoma pathophysiology.
152
III. NOVEL STRATEGIES FOR GLAUCOMA THERAPY: NEUROPROTECTION AND AXONAL REGENERATION
153
Neuroprotective strategies are extensively investigated in the context of several neurological
154
disorders, and form a therapeutic paradigm aiming at slowing down or preventing death of neurons.
155
In the case of glaucoma, where loss of RGCs progressively manifests over many years, a
156
neuroprotective therapy should enhance the survival of RGCs in the presence of chronic
157
stress/injury. Several approaches for neuroprotection in glaucoma are being investigated, targeting
158
neurotrophic withdrawal, excitotoxicity, oxidative stress, mitochondrial dysfunction, protein
159
misfolding, etc. (Baltmr et al. 2010). Given the importance of neurotrophins, such as e.g. brain
160
derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF) and nerve growth factor
161
(NGF), for the survival of RGCs (Weibel et al. 1995; Ji et al. 2004b), and the reduced axoplasmic
162
transport of these growth factors due to elevated IOP (Quigley et al. 2000), delivering neurotrophic
163
factors to the retina has been a key target in the development of neuroprotective therapies. Indeed,
164
multiple studies demonstrated the neuroprotective effect of BDNF, CNTF and NGF in animal models
165
of glaucoma (Johnson et al. 2011). These neurotrophic factor supplementation therapies are now
166
going into clinical trials for primary open angle glaucoma, e.g. trials with CNTF implants and NGF eye
167
drops (Chang and Goldberg 2012a), although the lack of methods for sustained, safe delivery of
168
biologically active neurotrophins hampers the broad clinical translation of this potential therapy.
169
Alternatively, neuroprotective agents preventing excitotoxicity, i.e. RGC apoptosis due to excessive
170
extracellular glutamate, have been investigated. This resulted in one of the only two clinical trials
171
evaluating neuroprotective agents for the treatment of glaucoma thus far. Indeed, memantine, a N-
172
methyl-D-aspartate (NMDA) receptor antagonist, prevents prolonged influx of Ca2+ ions through the
173
NMDA receptor channel and as such directly protects RGCs against excitotoxicity (Danesh-Meyer
174
2011). The other clinical trial, evaluating the use of the selective α2 receptor agonist brimonidine,
175
was based on the observation that this IOP lowering drug may protect RGCs from glaucomatous
7
176
neurodegeneration via a not yet known, IOP-independent mechanism (Lai et al. 2002; Dong et al.
177
2008). Nevertheless, despite good preclinical results, both clinical trials failed to deliver adequate
178
evidence for the applicability of memantine and brimonidine in the clinic, probably due to a too
179
narrow neuroprotective window of these agents (Weinreb 2007; Osborne 2009; Krupin et al. 2011).
180
Promoting survival of RGCs is a critical step in the development to a neuroprotective
181
strategy. However, in glaucoma also axons of RGCs are damaged, and as such, an ideal
182
neuroprotective glaucoma therapy should not only stimulate RGC survival but should also stimulate
183
axon regeneration (Chang and Goldberg 2012b). Clinical applications of the latter, however, seem to
184
be out of reach for now. For over three decades, the optic nerve crush (ONC) paradigm has been
185
used to study axonal regeneration of the optic nerve, first by stimulation of axonal outgrowth by
186
intraocular neurotrophic factor injection and peripheral nerve graft transplantation, and more
187
recently, by induction of controlled intraocular inflammation via experimental lens injury or
188
intravitreal zymosan injection. Moreover, manipulation of specific intracellular signaling pathways
189
has led to the identification of STAT3 (signal transducer and activator of transcription 3) and mTOR
190
(mammalian target of rapamycin) as central regulators in axonal outgrowth (Pernet and Schwab
191
2014). Despite these advances, it remains a challenge to induce sufficient axonal regeneration in
192
order to reinnervate retinal target areas in the brain, and restore visual function (de Lima et al. 2012;
193
Luo et al. 2013).
194
translatable to clinical practice.
Moreover, the current experimental treatments in rodents are not readily
195
Of note, ‘vasoprotection’ could be added as an additional therapeutic approach to prevent
196
glaucomatous neurodegeneration (Wierzbowska et al. 2010), focusing on the prevention of ischemia
197
and reperfusion injury. Indeed, in a subset of patients, optic neuropathy is a consequence of vascular
198
dysregulation, due to e.g. systemic circulatory disturbances and perfusion deficits of cerebral,
199
retrobulbar and ocular vessels (Wierzbowska et al. 2010; Yanagi et al. 2011). Besides neuroprotective
200
drugs targeting excitotoxicity, these patients would greatly benefit from therapies restoring e.g.
8
201
vascular tone, endothelial function and neurovascular coupling (Flammer et al. 2002a; Yanagi et al.
202
2011).
203
IV. THE RHO-ROCK PATHWAY
204
Rho is a member of the cytoplasmic Rho family of small GTP-binding molecules, which
205
belongs to the Ras-superfamily of GTPases. The Rho subfamily contains 3 isoforms (RhoA, RhoB and
206
RhoC) that are ubiquitously expressed. To become activated, Rho must be targeted to the membrane
207
via posttranslational modification (Bishop and Hall 2000). The activity of the Rho pathway is
208
regulated by signaling input from several growth factors, cytokines, mechanical stretch and the ECM,
209
and as such mediated by different classes of cell surface receptors (Etienne-Manneville and Hall
210
2002). Rho GTPases function as molecular switches and cycle between an inactive GDP and an active
211
GTP-bound state. Activation is regulated by guanine nucleotide-exchange factors, which catalyze the
212
exchange of GDP to GTP. On the other hand, GTPase activating proteins inhibit downstream signaling
213
by stimulating the hydrolysis of GTP to GDP (Bos et al. 2007) (figure 2). The Rho-associated coiled-
214
coil-containing protein kinases (also known as ROCK) are the first identified and most extensively
215
studied downstream effectors of Rho. ROCKs are essentially distributed in the cytoplasm, but are
216
translocated to the plasma membrane after RhoA activation (Matsui et al. 1996). In mammals, ROCKs
217
exist as two isoforms: ROCKI and ROCKII. ROCKs contain three domains, the catalytic kinase domain,
218
which is located at the N-terminus, followed by a coiled-coil region containing the Rho-binding site
219
and a pleckstrin-homology domain with a cysteine-rich repeat domain at the carboxyl terminus
220
(Riento et al. 2003). The two ROCK isoforms share an overall homology in their sequences of 65%.
221
The highest similarities are found in their kinase domains, which are 92% identical. Both ROCK
222
isoforms are ubiquitously expressed in most tissues. However, there is a significant difference in
223
tissue distribution. Higher levels of ROCKII are found in brain and muscles, whereas ROCKI is
224
predominately expressed in non-neuronal tissues such as liver and lungs (Nakagawa et al. 1996).
9
225
1. Downstream effectors of ROCK: well-known cytoskeletal regulators
226
Once ROCK is activated by GTP-bound Rho, it is able to phosphorylate multiple substrates,
227
which almost all play a critical role in regulating cytoskeletal dynamics (Figure 2). The best-
228
characterized substrate of ROCK is the cytoskeletal regulator myosin light chain (MLC).
229
Phosphorylation of MLC results in stimulation of actin-myosin interactions, which play a role in
230
several cytoskeletal functions like cell morphology, adhesions, motility and smooth muscle cell
231
contraction. ROCK also regulates MLC activity via the inhibition of MLC phosphatase (Leung et al.
232
1996; Kaneko-Kawano et al. 2012). The actin-binding kinases, LIM kinase (LIMK) 1 and 2, are other
233
downstream targets of ROCK and are involved in the regulation of actin filament stabilization. Active
234
ROCK phosphorylates LIMK and thereby enhances its ability to phosphorylate cofilin, an actin-binding
235
and depolymerizing factor that regulates the turnover of actin filaments. Phosphorylation of cofilin
236
by LIMK inactivates its activity. Therefore, the phosphorylation of LIMK by ROCK inhibits cofilin-
237
mediated actin filament disassembly and leads to an increase in the number of actin filaments
238
(Ohashi et al. 2000; Sumi et al. 2001). ROCK also affects the ezrin/radixin/moesin (ERM) family, a
239
family of proteins that promote cross-linking of integral membrane proteins with actin filaments near
240
the cell surface. Phosphorylation and activation of the ERM proteins by ROCK regulates membrane-
241
actin interactions, permitting cytoskeletal reorganization and the establishment of focal adhesions
242
between the actin cytoskeletal and ECM (Tsukita and Yonemura 1997; Shaw et al. 1998). Another
243
ROCK effector is adducin, a filamentous-actin-binding protein which promotes actin network
244
assembly beneath the cell membrane and plays a role in cell motility (Matsuoka et al. 2000).
245
Intermediate filament proteins such as vimentin, glial fibrillary acidic protein (GFAP) and
246
neurofilaments, are cytoskeletal structures important for maintaining cell integrity and mechanical
247
strength, and their phosphorylation by ROCK causes intermediate filament disassembly and is
248
needed in specific events such as cytokinesis (Riento and Ridley 2003). ROCKs are also important
249
regulators of the process of inflammation, by the activation of NF-κβ, which subsequently controls
10
250
the transcription of proinflammatory factors such as interleukins and tumor necrosis factor-α, in
251
different inflammatory cells (Doe et al. 2007; He et al. 2008).
252
2. The ROCK signaling cascade and its involvement in neuronal cell death and axonal outgrowth
253
Several other downstream components of the ROCK pathway are highly expressed in the
254
CNS, and are regulators of several neural processes, ranging from neuronal survival to axonal
255
outgrowth and guidance (Schmandke and Strittmatter 2007) (Figure 2).
256
ROCK is a known negative regulator of neurite extension, resulting in axon retraction and
257
growth cone collapse during development and axon regeneration (Gallo 2006; Pasterkamp and
258
Verhaagen 2006). Indeed, ROCK is an important regulator of collapsin response mediator protein-2
259
(CRMP-2) that plays a role in semaphorin-3A-mediated repulsive axon guidance (Arimura et al. 2000).
260
Other downstream targets that play a major role in neurite extension are microtubule-associated
261
proteins (MAPs), such as Tau or MAP-2, which are important players in neuronal morphology by
262
altering microtubule dynamics (Amano et al. 2003). One of the most recently discovered and
263
interesting downstream targets of ROCK is the phosphatase and tensin homolog (PTEN). ROCK
264
phosphorylates and subsequently activates PTEN, thereby negatively regulating neuronal survival
265
and axonal regeneration via the PIP3/Akt/mTOR pathway (Li et al. 2005; Tonges et al. 2011).
266
Neurofilaments are intermediate proteins found in high concentrations along axons of vertebrate
267
neurons. Activation of these proteins by ROCK results in intermediate filament disassembly and as
268
such alters axon integrity and axonal growth dynamics (Hashimoto et al. 1998; Walker et al. 2001).
269
GFAP is another intermediate filament protein that is highly expressed in astrocytes and has been
270
shown to be important in repair after CNS injury and the formation of a glial scar (McKeon et al.
271
1991). Noteworthy, the Rho-ROCK pathway can be activated by several myelin-associated molecules
272
that inhibit axonal regeneration in the CNS, such as myelin-associated glycoprotein (MAG), neurite
273
outgrowth inhibitor (Nogo) and oligodendrocyte myelin glycoprotein (OMgp) (Hasegawa et al. 2004;
274
Mimura et al. 2006; Kubo et al. 2008). Indeed, the inability of mature CNS neurons to regenerate
275
injured axons has been attributed to a loss of an inherent growth potential of cells and to inhibitory
11
276
signals associated with myelin and the glial scar. These myelin-associated inhibitory molecules all
277
bind with high affinity the Nogo receptor (NgR) (McKerracher and Winton 2002). Interaction of this
278
receptor with the extracellular domain of p75NTR is known to activate Rho GTPase, and as such
279
activate ROCK, in turn resulting in neurite outgrowth inhibition and growth cone collapse (Kaplan and
280
Miller 2003; Yamashita and Tohyama 2003). Chondroitin sulfate proteoglycans (CSPGs), such as
281
brevican, aggrecan, versican, phosphocan and neuroglycan-2, are upregulated by reactive astrocytes
282
in the glial scar, formed around the lesion site. These CSPGs constitute an important family of
283
inhibitory ECM molecules (Silver and Miller 2004). It has been proposed that these CSPGs interact
284
with the ROCK pathway, resulting in growth inhibition (Gungabissoon and Bamburg 2003; Monnier et
285
al. 2003; Gopalakrishnan et al. 2008).
286
Thus, many of the known or suggested upstream and downstream signaling molecules of the
287
ROCK pathway in the CNS are related to axonal outgrowth after CNS damage, which subsequently
288
implicates the major contribution of the ROCK pathway to the restricted axonal regeneration in the
289
adult mammalian CNS. Blocking the Rho-ROCK signaling pathway is therefore yet another new
290
strategy to promote axonal regeneration and repair after injury in the CNS.
291
V. ROCK INHIBITION AS A MULTI-TARGET APPROACH FOR GLAUCOMA TREATMENT
292
Given the important role of Rho and ROCK in different cellular processes, targeting this
293
pathway may be of high value for various diseases. As such, inhibition of ROCK has already been
294
investigated as a potential strategy for cardiovascular diseases (Shi and Wei 2013), bronchial asthma
295
(Kume 2008) and cancer (Itoh et al. 1999). More recently, ROCK has also been highlighted as a
296
promising target for neurological disorders, as inhibition of ROCK seems to promote neuronal cell
297
survival and axonal regeneration (Mueller et al. 2005; Fujita and Yamashita 2014).
298
1. ROCK inhibitors as IOP lowering and anti-scarring agents in glaucoma
299
IOP lowering drugs currently in clinical use for glaucoma, suppress the production of AH or
300
enhance uveoscleral outflow. None of them directly targets and/or improves conventional outflow
12
301
via the TM. Therefore, agents that increase AH outflow by modifying the TM are promising targets
302
for the development of a new IOP lowering strategy to treat most forms of glaucoma (Llobet et al.
303
2003). The cells of the TM possess smooth muscle-like properties, as evidenced by the expression of
304
α-smooth muscle actin (α-SMA) (de Kater et al. 1990), and their contraction-relaxation status has
305
been reported to influence AH outflow facility (Wiederholt et al. 2000). Interestingly smooth muscle
306
cell contraction is regulated predominately by the phosphorylation status of MLC, a main
307
downstream target of ROCK.
308
In vitro and ex vivo studies have reported Rho and ROCK expression in healthy and
309
glaucomatous TM cells (Honjo et al. 2001a; Rao et al. 2001; Nakajima et al. 2005; Goldhagen et al.
310
2012). Cellular effects and IOP-lowering efficacy of several ROCK inhibitors, such as HA-1077, H-
311
1152P, K-115 Y27632, Y-39983 and AMA0076 have been previously reported (Honjo et al. 2001a;
312
Honjo et al. 2001b; Rao et al. 2001; Waki et al. 2001; Tokushige et al. 2007; Lu et al. 2008; Fukunaga
313
et al. 2009; Nishio et al. 2009; Isobe et al. 2014; Van de Velde et al. 2014a), and indeed unveiled that
314
ROCK regulates AH outflow by directly affecting contractility and cellular organization of the TM cells
315
(Honjo et al. 2001a; Rao et al. 2001). Overall, ROCK inhibition induces relaxation, actin cytoskeletal
316
reorganization and loss of cell adhesions of TM cells, leading to better drainage of AH trough the TM.
317
Not all patients respond adequately to anti-glaucoma medication making invasive therapies,
318
such as laser trabeculoplasty and/or filtration surgery, necessary for the treatment of glaucoma (Lee
319
and Higginbotham 2005). Unfortunately, filtration surgery frequently fails as the result of excessive
320
postoperative wound healing and subsequent scarring, leading to poor postoperative IOP control
321
and, as a consequence, progression of visual field loss (Addicks et al. 1983). During scar formation,
322
the cytokine TGF-β induces a contractile response in fibroblasts, which subsequently differentiate
323
into myofibroblasts. In vitro studies have shown that ROCK inhibitors might prevent this
324
transdifferentiation, probably via decreasing cell contraction, which implies that ROCK inhibitors may
325
reduce postoperative scarring after glaucoma filtration surgery (Meyer-ter-Vehn et al. 2006; Zhou et
326
al. 2013). Indeed, Honjo et al. showed that the ROCK inhibitor Y-27632 inhibited fibroproliferation
13
327
and collagen deposition in an animal model of glaucoma filtration surgery, leading to a prolonged
328
survival of the postoperative bleb (Honjo et al. 2007).
329
Thus, ROCK inhibition may possess therapeutic potential as a novel IOP lowering strategy and
330
be able to reduce postoperative scarring after filtration surgery, opening new perspectives for a
331
more efficient IOP control.
332
2. ROCK inhibitors as novel neuroprotective and axon regenerative agents in glaucoma
333
However, in NTG patients, glaucomatous damage occurs despite normal IOP levels and also
334
clinical evidence indicates that IOP lowering therapy does not prevent progression in all patients
335
(VanVeldhuisen et al. 2000). Since glaucomatous optic neuropathy eventually results in progressive
336
loss of RGCs, it is reasonable to expect that a neuroprotective therapy may be of high value as an
337
additional treatment modality for glaucoma. Such a therapy should not only directly promote RGC
338
survival, but should also encourage axonal regeneration, thereby recovering functional connections
339
(Chang and Goldberg 2012b). As we will discuss further in this review, there is increasing evidence
340
that ROCK is upregulated during glaucomatous damage and consequently, that ROCK inhibition
341
directly protects RGC survival and promotes axon regeneration. This emphasizes that ROCK inhibition
342
may be a versatile strategy for the treatment of glaucoma.
343
Much of the evidence concerning ROCK inhibition as a novel neuroprotective therapy has
344
been elucidated in glaucoma models (Kitaoka et al. 2004; Hirata et al. 2008; Lingor et al. 2008; Tura
345
et al. 2009; Alt et al. 2013). Furthermore, it has also been demonstrated that inhibition of ROCK limits
346
neurodegeneration in several in vitro, ex vivo and in vivo animal models for e.g. Amyotrophic Lateral
347
Sclerosis (ALS) (Tonges et al. 2014), Multiple Sclerosis (MS) (Zhang et al. ; Li et al. 2014a),
348
Huntington's (Li et al. 2009), Parkinson's (Tonges et al. 2012; Borrajo et al. 2014; Labandeira-Garcia et
349
al. 2014) and Alzheimer's disease (Mueller et al. 2005; Herskowitz et al. 2013; Song et al. 2013),
350
stroke/ischemia (Toshima et al. 2000; Satoh et al. 2001; Rikitake et al. 2005; Ding et al. 2009;
351
Koumura et al. 2011) and Spinal Muscular Atrophy (Bowerman et al. 2012; Froger et al. 2012). These
14
352
prevalent neurodegenerative disorders, which are characterized by a progressive loss of specific
353
neurons/axons, thereby leading to important dysfunctional consequences, still remain largely
354
incurable. Therefore, it is becoming increasingly clear that a therapeutic strategy with a
355
neuroprotective effect has an enormous future potential as effective treatment (Bull and Martin
356
2007; Cheung et al. 2008).
357
In the next paragraphs, this review will provide a detailed overview of the current knowledge
358
related to the potential of ROCK inhibition and the rationale why ROCK targeting seems a promising
359
tool to ensure neuronal protection and/or axonal regeneration.
360
2.1. ROCK expression in the healthy and injured/diseased CNS
361
The amount of studies describing ROCKI/II expression in the neuroretina is rather poor. In the
362
healthy rat retina, RhoA, ROCKI and ROCKII expression were reported to co-localize mainly in the
363
retinal microvessels (Arita et al. 2009). Another study could not demonstrate immunopositivity for
364
ROCKII in healthy rat retinas. However, axotomy of the rat optic nerve induced ROCKII expression,
365
but only in the RGC layer (RGCL), suggesting that ROCK might be detrimental to RGC survival and/or
366
contribute to a regeneration-inhibitory cascade (Lingor et al. 2008). Immunostainings for RhoA in rat
367
retinas also showed, after injection with the glutamate analog N-methyl-D-aspartate (NMDA), an
368
increased immunopositivity in the RGCL, the inner plexiform layer (IPL) and some somata in the inner
369
nuclear layer at 1 day post injection (1 dpi). Western blot data confirmed these findings and
370
demonstrated an upregulation of ROCKII in the NMDA-treated retina at 1 dpi, thereby suggesting a
371
commitment of Rho-ROCK signaling in NMDA/glutamate-induced retinal excitotoxicity (Kitaoka et al.
372
2004). Rho and ROCK expression in the ONH of glaucomatous patients is also significantly increased
373
as compared to healthy controls, indicating a possible involvement of the Rho-ROCK pathway in the
374
pathophysiology of optic nerve damage in glaucoma (Goldhagen et al. 2012). Clearly, additional
375
studies are necessary to investigate the cellular localization of ROCK, as well as its expression
376
patterns and activity levels in the healthy and glaucomatous retina, in order to understand the
15
377
inter/intracellular mechanisms contributing to RGC death and neurite outgrowth inhibition after
378
injury.
379
Studies in the healthy and diseased/injured brain and spinal cord reveal expression of ROCKII,
380
and to a much lower extent ROCKI, in neurons, endothelial cells and microglia (Nakagawa et al. 1996;
381
Deyts et al. 2009; Duffy et al. 2009). Moreover, treatment of PC12 cells with Nogo, a myelin-
382
associated neurite outgrowth inhibitor, did not only increase ROCKII activity, but also resulted in a
383
translocation of ROCKII towards the membrane (Alabed et al. 2006). This finding indicates that
384
ROCKII is activated by Nogo and strengthens the potential of ROCK inhibition in stimulating axonal
385
regeneration.
386
2.2. ROCK inhibition as a promising tool to induce axon regeneration in the injured visual system
387
An effective treatment to enable damaged axons to regrow beyond the lesion site and
388
reform functional connections should ideally require interventions targeting the cell-intrinsic growth
389
capacity, as well as the extrinsic inhibitory environment. In general, much progress concerning the
390
cellular and molecular mechanism underlying the failure of adult axons to regenerate, has been
391
made using studies in the rodent optic circuit. Functional recovery in the visual system implies the
392
proper regrowth of damaged axons to reinnervate the lateral geniculate nucleus and the superior
393
colliculus with sufficient numbers (Pernet et al. 2013; Murray 2014). Studies combining pten deletion
394
with either ‘suppressor of cytokine signaling 3’ (socs3) deletion, or zymosan and CPT-cAMP
395
injections, have demonstrated the potential of RGCs to reactivate their intrinsic axon growth
396
program and regenerate the full length of the optic nerve, across the optic chiasm (de Lima et al.
397
2012; Luo et al. 2013). Indeed, complete axonal regeneration to the lateral geniculate nucleus and
398
superior colliculus, along with partial recovery of visual functions, was observed upon pten deletion
399
plus CTP-cAMP and zymosan administration (de Lima et al. 2012). In an alternative approach,
400
combining pten and socs3 co-deletion with CNTF supplementation, short-distance axonal
401
regeneration to the suprachiasmatic nucleus was seen, yet no reinnervation of long-distance target
402
areas (Luo et al. 2013). Notably, adult RGCs were shown to be able to reactivate de novo axon growth
16
403
upon pten and socs3 co-deletion after injury, thereby making a future therapy to promote axon
404
repair more and more feasible. Yet, the poor number of RGC axons reaching their long-distance
405
target areas and the significant axon misguidance in the optic nerve and brain underscore the need
406
for further research efforts.
407
In vitro application of the ROCK inhibitors Y-27632, fasudil (HA-1077) and dimethylfasudil (H-
408
1152) on Ntera-2 neurons, PC12 cells and primary dorsal root ganglion cells, grown on a CSPG or
409
myelin substrate, was shown to overcome inhibition of neurite outgrowth (Fournier et al. 2003;
410
Zhang et al. 2006; Lingor et al. 2007; Cheng et al. 2008; Gopalakrishnan et al. 2008). It has also been
411
demonstrated in vivo that the amount, thickness and/or length of axons is increased after sciatic
412
nerve crush and spinal cord injury, and even locomotor activity, is improved after application of
413
ROCK inhibitors (Fournier et al. 2003; Sung et al. 2003; Hiraga et al. 2006; Cheng et al. 2008; Gunther
414
et al. 2014). Recently, inhibition of ROCK has proven to inhibit demyelination in an experimental
415
autoimmune encephalomyelitis animal model for MS (Gao et al. 2013).
416
As such, the potential of ROCK inhibitors to support axonal outgrowth and regeneration has
417
increased during the last years. Importantly, much evidence has been obtained from in vitro and ex
418
vivo studies on RGCs and retinal explants, and from experimental in vivo glaucoma models.
419
2.2.1. The neurite outgrowth-potential of ROCK inhibition in in vitro and ex vivo models for glaucoma
420
Addition of Y-27632, or the more potent Y-39983, to RGC-5 cells or postnatal rat primary
421
RGCs grown on a permissive substrate, demonstrated a clearly induced neurite outgrowth. Neurite
422
outgrowth was even more pronounced after combining Y-27632 with CNTF on these 2 cell cultures
423
(Lingor et al. 2008; Bermel et al. 2009; Tokushige et al. 2011) (Table 1). CNTF exerts its main
424
biological functions via activation of the Jak/STAT3, PI3K/Akt and MAPK/ERK signaling pathways (Park
425
et al. 2004), which are, interestingly, also activated upon ROCK inhibition (Lingor 2008, Pernet 2013,
426
Koch 2014). In general, STAT3 has been considered a central regulator of growth and regeneration of
427
adult RGCs and to promote their intrinsic growth capacity via transcription of growth-associated
428
genes and stabilization of the RGC cytoskeleton (Ng et al. 2006; Selvaraj et al. 2012; Pernet and
17
429
Schwab 2014). In contrast, STAT3 was once suggested to act as a negative regulator of neurite
430
outgrowth, proposing that Y-27632 may further increase neurite elongation in combination with
431
CNTF, by inhibiting STAT3 phosphorylation (Lingor et al. 2008). Furthermore, ROCK inhibition also
432
results in suppression of Lim kinase (LIMK) phosphorylation, which is induced upon binding of
433
myelin-derived inhibitory ligands (MAG, Nogo, OMgp) to their NgR receptor and leads to neurite
434
outgrowth inhibition and growth cone collapse (Sagawa et al. 2007). Similarly, ROCK inhibition can
435
relief the repulsive effects of glial-derived inhibitory ligands, such as CSPGs (Monnier et al. 2003).
436
Indeed, treatment of postnatal rat RGCs, grown on an inhibitory CSPG substrate, with Y-27632
437
significantly induced neurite length as compared to controls (Bermel et al. 2009). On the other hand,
438
treating adult rat retinal cells with a panel of inhibitory molecules, including Nogo-A, MAG, OMgp
439
and CSPG, prevented the stimulatory effect of Y-27632 on RGC neurite outgrowth (Ahmed et al.
440
2009) (Table 1). These, at first sight contradictory, findings might be explained by the age at which
441
the RGC cultures were obtained and might result from the suppressed 'growth activated state' of
442
adult CNS neurons, due to e.g. reduced cAMP levels, and as such, a limited regenerative capacity (Cai
443
et al. 2001). Remarkably, stimulation of adult retinal cells with Y-27632, in combination with CNTF,
444
significantly enhanced the number and length of RGC neurites, above the levels of CNTF treatment
445
alone. As such, Y-27632 does not seem to support neurite outgrowth of disinhibited adult RGCs, but
446
is able to enhance CNTF-induced neurite outgrowth (Ahmed et al. 2009). Similar findings were
447
observed when Y-27632-treated adult RGCs were supplemented with forskolin, which induces cAMP
448
levels. The combination of the three compounds enhanced neurite outgrowth of adult RGC neurites
449
to even greater levels than combined Y-27632 and forskolin treatment (Ahmed et al. 2009).
450
As the currently existing ROCK inhibitors do not distinguish between ROCKI and ROCKII, and
451
have been described to also block several other kinases and neuronal receptors (Mueller et al. 2005),
452
Koch et al. circumvented this issue by generating shRNA-expressing adeno-associated viral (AAV)
453
vectors to specifically downregulate ROCKII. Transduction of postnatal rat RGCs with AAV.ROCK2-
454
shRNA showed a trend towards longer neurites when grown on the permissive laminin substrate.
18
455
Moreover, knockdown of ROCKII enabled a full rescue of the growth-inhibiting effect of CSPG on
456
neurite outgrowth (Koch et al. 2014).
457
When embryonic rat retinal explants were applied in a stripe assay, consisting of alternate
458
laminin-coated and laminin/CSPG-coated stripes, or on a uniform laminin/CSPG substrate, axons
459
tended to avoid the CSPG substrate. Addition of Y-27632 clearly reduced this repulsive property and
460
significantly increased the amount and length of the outgrowing axons in comparison to non-treated
461
explants (Monnier et al. 2003) (Table 1). Administration of Y-27632 on adult rat retinal explants in
462
culture, dissected after optic nerve crush (ONC) (Herskowitz et al.), showed an increased number of
463
outgrowing neurites. When Y-27632 was applied together with Indolinone A, an inhibitor of cyclin
464
dependent kinase 5 (cdk5), which triggers a cascade of neurotoxic pathways in many
465
neurodegenerative disorders, an additional increase in outgrowth could be observed (Bermel et al.
466
2009). Supplementation of Y-27632 or Y-39983 also significantly induced neurite extension in adult
467
cat retinal explant studies (Sagawa et al. 2007; Ichikawa et al. 2008) (Table 1). Of note, the number of
468
extending neurites was higher when applying Y-39983, even at lower concentration than Y-27632,
469
thereby confirming the higher potency of Y-39983. Yet, no difference in neurite length could be
470
observed. Importantly, both Y-compounds also promoted the extension of glial processes in the cat
471
retinal explant assay (Sagawa et al. 2007). Notably, application of fasudil on adult cat retinal tissue
472
explants only induced glial fiber extension, while no neurite-outgrowth promoting effect was
473
observed (Ichikawa et al. 2008).
474
Using a retinal explant model, recently described by Buyens et al. (Buyens et al. 2014), our
475
group was able to demonstrate a clear neurite outgrowth-promoting effect of Y-27632 and Y-39883
476
in postnatal mouse retinal explants, respectively a 2 and 2.5 fold increase as compared to control
477
explants (Figure 3a) (Gaublomme et al. 2013). Also here, no significant neurite extension-promoting
478
effect was observed for any of the tested ROCK inhibitors (Figure 3b). Nevertheless, we also observed
479
a stimulating effect on glial process outgrowth (not shown). It is known that reactive glia, being
480
Müller glia and astrocytes, are present in the glaucomatous retina, although their exact role in the
19
481
disease pathogenesis seems to be ambiguous (Nickells 2007; Johnson and Morrison 2009). However,
482
their function as mediators of RGC survival and regeneration is being strengthened during the last
483
years and, amongst others, supported by their production of glial-derived growth factors (Muller et
484
al. 2007; Leibinger et al. 2009; Lorber et al. 2009). Next to soluble factors, also the expression of
485
membrane-bound glial derived growth factors, as well as axonal guidance molecules, contribute to
486
the, at least in part, RGC growth-promoting effect of activated retinal glia (Lorber et al. 2012).
487
These in vitro and ex vivo findings indicate that ROCK inhibitors have a strong capacity to
488
support axonal outgrowth, even in the presence of growth-inhibitory molecules, which mimics the in
489
vivo situation after axonal damage.
490
2.2.2. ROCK inhibitors promote in vivo axonal regeneration after optic nerve traumatic lesions
491
Intravitreal injection of Y-27632 or Y-39983 in the adult cat or rat, immediately after ONC,
492
promoted the number and length of regenerating axons passing the crush site, as compared to
493
crushed control eyes (Table 1). Also intravitreal injection of AAV.ROCK2-shRNA significantly increased
494
the number and length of growth associated protein 43 (GAP43)-positive neurites in the rat optic
495
nerve, after ONC, as compared to an AAV.EGFP-shRNA control vector (Koch et al. 2014). The
496
combined application of Y-27632 and the cdk5 inhibitor Indolinone A, resulted in a significantly
497
higher number, rather than an increased length, of regenerating axons beyond the ONC site, in
498
comparison to either compound alone (Bermel et al. 2009). On the other hand, administration of
499
different doses of fasudil or dimethylfasudil in a similar experimental set-up, revealed only minor
500
effects on axonal outgrowth (Lingor et al. 2007; Bermel et al. 2009) (Table 1). Repeated intravitreal
501
injections of Y-27632 in a rat sciatic nerve graft model, in which the sciatic nerve is transplanted onto
502
the optic nerve stump, did not result in additional axons in the nerve graft as compared to PBS-
503
treated eyes (Lingor et al. 2008) (Table 1). As peripheral nerves lack the major myelin-associated
504
inhibitory molecules, such as Nogo-A, this semi-permissive model allows to study the effect of
505
compounds on the intrinsic regenerative capacity of RGCs, as RGCs show a spontaneous regeneration
506
response into the graft (Thanos et al. 1997). The reported data would then suggest that inhibition of
20
507
ROCK rather promotes axon regeneration by modulating the extracellular environment at the injury
508
site than by altering the intrinsic growth capacity of RGCs. Anyway, co-application of Y-27632 and
509
CNTF enhanced the number of regenerating axons into the graft, to a greater extent than CNTF
510
alone, again indicating an advantageous interplay for these 2 molecules (Lingor et al. 2008). A
511
stronger regenerative response after combined administration of both compounds, as compared to
512
Y-27632 or CNTF treatment alone, was also observed in the (non-permissive) ONC model in the rat,
513
with a clearly more pronounced effect on the number, rather than the length, of regenerating axons
514
(Lingor et al. 2008). Importantly, also repeated injections of Y-27632 alone increased the number of
515
regenerating axons past the lesion site more than two-fold in comparison to control animals, thereby
516
reflecting the potential of ROCK inhibitors to overcome inhibitory myelin substrates. The adult rat
517
sciatic nerve graft model has also been applied to study the axon regenerative potential of Y-33983
518
(Tokushige et al. 2011) (Table 1). In comparison to the findings of Lingor et al. with Y-27632 (Lingor et
519
al. 2008), intravitreal injection and sponge application of solely Y-39983, dose-dependently increased
520
the number of RGCs with regenerating axons in peripheral nerve-transplanted retinas, as compared
521
to saline-administered eyes. Although Y-39983 is more potent than Y-27632 and might as such
522
induce axonal outgrowth to a higher extent (without CNTF), these two studies are difficult to
523
compare, as the amount and length of regenerated axons in the optic nerve was not determined in
524
the latter study (Tokushige et al. 2011).
525
Using a mouse ONC model and intravitreal injection of STAT3 cDNA-expressing AAV vectors,
526
Pernet et al. demonstrated that intracellular activation of STAT3 enhances axonal regeneration
527
(Pernet et al. 2013). However, STAT3-mediated axon elongation is characterized by massive
528
directionality and guidance problems, most likely due to the presence of growth-inhibitory molecules
529
in the optic nerve environment. Interestingly, additional intraocular delivery of Y-27632 did not only
530
result in an increased amount of outgrowing neurites (and surviving RGCs), but also counteracted the
531
Rho-LIMK-mediated misguidance effects and normalized the morphology and trajectories of the
532
regenerating axons. Intravitreal application of Y-27632 alone was not able to significantly induce
21
533
axonal regeneration. Importantly, combined delivery of Y-27632 and the Müller glia-selective AAV
534
vector ShH10.DH-CNTF, resulted in glial CNTF expression and activation of STAT3 in RGCs, eliciting a
535
more robust axonal outgrowth after ONC as compared to single ShH10.DH-CNTF or combined STAT3
536
and Y-27632 treatment (Pernet et al. 2013). These findings again support the notion that
537
combinatorial strategies will be needed. Indeed, co-injection of Y-27632 and AAV2.STAT3 raised
538
phosphorylated STAT3 (pSTAT3) levels and expression of STAT3 target genes, such as GAP43, after
539
injury (Pernet et al. 2013).
540
In summary, ROCK(II) inhibition is recently brought forward as a potential strategy to help to
541
overcome restriction of axonal regrowth in the injured CNS, such as the visual system. The amount of
542
studies on the RGC axon regenerative potential of ROCK inhibitors in animal models of glaucoma is
543
clearly increasing over the last years. As such, it seems that ROCK inhibitors might induce the number
544
of outgrowing axons, and help in a proper guidance of the axons towards their target areas, albeit in
545
combination with other neuroprotective and/or regenerative molecules.
546
2.3. ROCK inhibition as a promising tool to support neuronal survival in the injured visual system
547
Several studies highlighting the regenerative potential of ROCK inhibitors, also reported a
548
beneficial effect of ROCK inhibition on neuronal survival. However, the mechanisms responsible for
549
the neuroprotective effects of ROCK inhibitors in experimental models of neurodegenerative
550
disorders, are only gradually being elucidated over the past years. Modulation of the PTEN/Akt and
551
MAPK/ERK signaling pathways (Lingor et al. 2008; Tonges et al. 2012), a decrease in oxidative stress,
552
enhanced expression of neurotrophic factors (Li et al. 2014b) and attenuation of microglial activity
553
(Zhao et al. 2006; Suzuki et al. 2007; Borrajo et al. 2014) and CNS inflammatory responses (Ding et al.
554
2010; Villar-Cheda et al. 2012; Borrajo et al. 2014; Li et al. 2014a; Tonges et al. 2014), have all been
555
put forward as possible mechanisms underlying the observed increase in neuronal survival.
556
2.3.1. In vitro and ex vivo studies on the neuroprotective potential of ROCK inhibitors
557
Lingor et al. (2008) demonstrated that addition of Y-27632 significantly increased survival
558
and decreased apoptosis of serum-deprived RGC-5 cells and, although to a lesser extent, of
22
559
neurotrophin-deprived primary rat RGCs (Table 2). Combination of Y-27632 with CNTF in vitro,
560
resulted in a synergistic effect on RGC survival. CNTF has been repeatedly described to protect RGCs
561
in vitro and in vivo, in animal models of traumatic optic neuropathies and ocular hypertension
562
(Lehwalder et al. 1989; Mey and Thanos 1993; Weise et al. 2000; Ji et al. 2004a; Maier et al. 2004;
563
Park et al. 2004; Zhang et al. 2005). The neuroprotective effect of Y-27632 seems to be associated
564
with upregulated levels of phosphorylated MAPK, which becomes remarkably more phosphorylated
565
or activated after combined administration with CNTF, indicating that inhibition of ROCK activates
566
MAPK downstream signaling pathways that are also used by CNTF. pAkt levels were not altered after
567
Y-27632, but showed a clear trend towards upregulation after combination treatment. Addition of
568
CNTF onto RGC-5 cultures resulted in increased pSTAT3 levels. However, application of the ROCK
569
inhibitor alone, as well as the combination of both substances, did not alter or even decreased
570
pSTAT3 levels compared to CNTF treatment alone, indicating that ROCK weakens some CNTF
571
signaling pathways, while amplifying others such as MAPK and Akt pathways (Lingor et al. 2008). Also
572
Bermel et al. demonstrated increased survival of growth factor-deprived primary rat RGCs in culture
573
when supplemented with Y-27632 (Bermel et al. 2009) (Table 2). Combined application of Y-27632
574
and the cdk5 inhibitor Indolinone A promoted RGC survival to a higher extent than treatment with
575
either substance alone. Furthermore, and in accordance to the findings of Lingor et al. (Lingor et al.
576
2008), administration of Y-27632 to serum-deprived RGC-5 cells resulted in a minor increase in
577
pMAPK levels (Bermel et al. 2009). These in vitro observations indicate that, next to the MAPK
578
signaling pathway, additional survival promoting mechanisms mediate the neuroprotective effect of
579
ROCK inhibition. Importantly, the origin and nature of the RGC-5 cell line has been the subject of
580
controversy during recent years, and it is now recognized that it is in fact of mouse photoreceptor
581
origin (Krishnamoorthy et al. 2013; Sippl and Tamm 2014). Nevertheless, it is still widely used as a
582
tool to follow-up initial hypotheses that require a transformed retina cell line of neuronal origin
583
(Sippl and Tamm 2014). It is therefore imperative that findings are subsequently tested with
584
complementary, biologically relevant tools such as primary RGCs or ex vivo and in vivo models
23
585
(Krishnamoorthy et al. 2013; Sippl and Tamm 2014). Indeed, all RGC-5 data mentioned in this review
586
has been confirmed in more relevant biological backgrounds, as can readily be appreciated from
587
tables 1 and 2.
588
In addition to these cell culture studies, the effects of ROCK inhibition on RGC survival were
589
also investigated using different ex vivo models. Administration of dimethylfasudil (H-1152P) to
590
serum-deprived organotypic cultures of adult mouse retinas, significantly and dose-dependently
591
reduced cell death in the RGC layer (Tura et al. 2009) (Table 2). In addition, macro- and microglial
592
reactivity, which was dramatically elevated after serum deprivation, was clearly attenuated by
593
dimethylfasudil, and corresponded to the observed reduction in the cytokines levels produced by
594
these reactive glial cells. The neuroprotective potential of dimethylfasudil was confirmed in an ex
595
vivo model with perfused bovine retinas under hypoxia (Alt et al. 2013). Similarly, macro- and
596
microglial activities were suppressed after dimethylfasudil application, as demonstrated e.g. by the
597
preservation of quiescent, ramified microglia. These ex vivo observations suggest that inhibition of
598
ROCK is able to protect RGCs from cytotoxicity, an effect that might be mediated through
599
suppression of glial reactivity.
600
To conclude, several in vitro and ex vivo findings already point to the existence of ROCK
601
downstream signaling pathways affecting RGC survival. As ROCK inhibitors seem to possess the
602
ability to enhance neuroprotective capacities of other molecules, more in-depth insight into the
603
underlying working mechanisms and molecular players is needed. In addition, the effect of ROCK
604
inhibition on glial reactivity should be further considered to broaden our knowledge about the
605
complex actions of micro- and macroglia in neurodegeneration.
606
2.3.2. ROCK inhibitors as RGC survival-promoting molecules in animal models for glaucoma
607
Because of the involvement of RhoA in NMDA receptor signaling (Norenberg et al. 1999;
608
Nakazawa et al. 2003), a role of RhoA and ROCK in NMDA-induced retinal neurotoxicity was
609
presumed (Kitaoka et al. 2004). Co-administration of NMDA and fasudil diminished NMDA-induced
610
cell loss in the RGCL and prevented the reduction in IPL thickness, as compared to single NMDA
24
611
injected retinas, thereby proposing that inhibition of ROCK is neuroprotective against glutamate-
612
related excitotoxicity (Kitaoka et al. 2004)(Table 2). Also in a transient retinal ischemia model in rats,
613
cell loss in the RGCL and reduction of IPL thickness was diminished after intravitreal injection of Y-
614
27632 (Hirata et al. 2008). Moreover, disturbance of vascular endothelial cell alignment and
615
leukocyte infiltration was dramatically suppressed in the post-ischemic retina after Y-27632
616
administration, and might underlie the observed diminished neuronal cell death. Lingor et al.
617
demonstrated a neuroprotective role for Y-27632 after repeated intravitreal injections in the adult
618
rat eye after complete transection of the optic nerve (Lingor et al. 2008). The neuroprotective
619
capacities of dimethylfasudil were shown in an ONC model in the rat, where pretreatment with
620
dimethylfasudil reduced RGC apoptosis in a dose-dependent matter (Herskowitz et al.). Remarkably,
621
also here an additional attenuation of macro- and microglial reactivity was observed (Tura et al.
622
2009). An increased RGC survival was also observed in a mouse ONC model after daily oral treatment
623
with either fasudil or K-115, a novel ROCK inhibitor with high selectivity for ROCKII, as compared to
624
vehicle-treated mice (Yamamoto et al. 2014). Moreover, the mRNA levels of the RGC markers Thy1.2
625
and Brn3a were increased after K-115 or fasudil treatment. K-115 likely attenuates oxidative stress
626
and as such, retard RGC death and glaucoma progression, by reducing the levels of oxidized lipids,
627
reactive oxygen species and NAPDH oxidase 1, known to be involved in the ONC-induced ROS
628
production pathway (Yamamoto et al. 2014). Koch et al. (2014) showed significantly enhanced RGC
629
survival in an ONC model in rats after retinal transduction with AAV.ROCK2-shRNA. In addition,
630
axonal degeneration on the proximal side of the crush was clearly retarded after ROCKII inhibition.
631
Part of the specific ROCKII-inhibitory effects on RGC survival and axonal de- and regeneration,
632
observed after AAV.ROCK2-shRNA transduction, could probably be assigned to decreased
633
intraneuronal activity of calpain and caspase-3 and increased levels of pAkt and CRMP-2 and
634
autophagic flux (Koch et al. 2014).
635
The fact that ROCK(II) inhibition limits RGC apoptosis and supports RGC survival in various
636
animal models of glaucoma, points to its broad window of action, which might be beneficial and even
25
637
mandatory for the treatment of a complex multivariate disorder like glaucoma. Nevertheless, the
638
neuroprotective effect of ROCK inhibition is modest (~10% survival increase), and much lower in
639
comparison to e.g. CNTF of BDNF trophic support.
640
3. ROCK inhibition as promising regulators of ocular blood flow
641
Risk factors of vascular nature, leading to disturbed ocular perfusion, have been positively
642
correlated with the appearance of glaucoma. In some patients, mostly patients with NTG, vascular
643
disorders even play a predominate role. Therefore, drugs that improve ocular blood flow to the ONH
644
are promising as an additional treatment modality for glaucoma.
645
Although not studied in any detail yet, ROCK inhibitors were recently shown to affect ocular
646
blood flow. Tokushige et al. showed that topical administration of Y-39983 significantly increased
647
blood flow at the ONH in rabbit eyes (Tokushige et al. 2011). In another study, intravenous infusion
648
or topical treatment with fasudil suppressed the impairment and even improved ONH blood flow,
649
induced by a nitric oxide synthase inhibitor or endothelin-1 in rabbits (Sugiyama et al. 2011). These
650
blood flow-improving effects of ROCK inhibitors can be explained by the fact that the Rho-ROCK
651
pathway is involved in MLC regulated contraction of smooth muscle cells surrounding blood vessels
652
in the ONH. Indeed, activation of MLC is regulated by the counteracting enzymes MLC kinase and
653
MLC phosphatase and the activity of MLC phosphatase is inhibited by ROCK causing smooth muscle
654
cell contraction (Uehata et al. 1997).
655
Considering the role of vascular dysregulation and impaired ocular perfusion in the
656
pathogenesis of glaucoma, and especially in patients with NTG, these data indicate an additional
657
advantage of ROCK inhibitors as possible vasoprotective drugs for the treatment for glaucoma.
658
V. FUTURE PERSPECTIVES: (PRE-)CLINICAL DEVELOPMENT OF ROCK INHIBITORS FOR GLAUCOMA TREATMENT
659
Given the important role of ROCK signaling in numerous cellular processes, ROCK inhibitors
660
are of potential interest for the treatment of a variety of cardiovascular, inflammatory, autoimmune
661
and neurodegenerative diseases (Doe et al. 2007; Fernandes et al. 2007; Kast et al. 2007; Liao et al.
26
662
2007; LoGrasso and Feng 2009). Specifically for neurodegenerative diseases, spinal cord injury, optic
663
nerve injury, traumatic brain injury and stroke, Alzheimer’s, Parkinson’s and Huntington’s disease,
664
amyotrophic lateral sclerosis and multiple sclerosis have been named as potential indications.
665
Indeed, the improved neurological recovery after spinal cord lesion observed in a phase I/IIa clinical
666
trial with BA-210 (Cethrin, BioAxone) (Fehlings et al. 2011), illustrates that RhoA/ROCK inhibitors are
667
a rational therapeutic approach for the treatment of neurological disorders. Nevertheless, today only
668
one ROCK inhibitor, fasudil (Eril, Asahi Kasei Pharma), has been approved in Japan for the treatment
669
of cerebral vasospasm (Zhao et al. 2006; Suzuki et al. 2007). It is generally believed that this limited
670
success of clinical applications of ROCK inhibitors is due to their profound effect on blood pressure
671
(Kast et al. 2007). As a consequence, avoiding systemic exposure, recent development of ROCK
672
inhibitors has been largely restricted to topical applications, e.g. clinical trials with Y-39983
673
(Novartis), K-115 (Kowa Company), AR-12286, AR-13324 (Aerie Pharmaceuticals) and ATS907
674
(Altheos), to evaluate their IOP lowering effect. However, even when applied topically to the eye,
675
side effects such as conjunctival hyperemia are seen (Tokushige et al. 2007; Tanihara et al. 2008;
676
Mandell et al. 2011). Overall, most available ROCK inhibitors have a narrow therapeutic window, due
677
to adverse effects resulting from either their low specificity or their interference with off-target
678
ROCK-dependent cellular processes. While this limited specificity of current ROCK inhibitors, i.e.
679
inhibition of other protein kinases and lack of specificity for ROCK isoforms (Chen et al. 2011; Shi et
680
al. 2013), remains a concern, so-called ‘soft’ ROCK inhibitors (Amakem Therapeutics) have been
681
developed to overcome the second issue. These compounds, structurally related to Y-27632, are
682
locally acting drugs that are designed to be relatively stable at the site of action and undergo
683
metabolic inactivation by controlled conversion into a predictable, non-functional metabolite (Bodor
684
and Buchwald 2008). As a result, off-target activity is avoided and these compounds thus display a
685
unique pharmacokinetic profile and a superior safety profile (Boland et al. 2013). Indeed, topical
686
administration of AMA0076 potently lowered IOP in New Zealand white rabbits with minimal
687
hyperemia (Van de Velde et al. 2014b). Of note, as ROCKI and ROCKII have very distinct expression
27
688
profiles (Chen et al. 2011; Shi et al. 2013), the local mode of action of these soft ROCK inhibitors
689
implies that unwanted inhibition of the second ROCK isoform is dramatically reduced. In addition,
690
alternative pharmacological variants/classes are being generated and tested in various experimental
691
disease models, in order to improve kinase selectivity, cellular activity, microsomal stabilization and
692
pharmacokinetic properties of ROCK inhibitors (Kubo et al. 2008; Tonges et al. 2011).
693
VI. CONCLUSION
694
Since the adult mammalian CNS lacks the capacity to replace lost nerve cells or to regenerate
695
injured axons, neurodegenerative and neurotraumatic disorders seriously affect life quality and
696
human wellbeing. Also glaucoma, which is increasingly being recognized as a multifactorial disease, is
697
characterized by loss of functional RGCs, causing progressive visual field loss. IOP lowering therapies
698
still remain the mainstay of glaucoma therapy. However, considering IOP elevation as the only
699
treatable risk factor, is increasingly being challenged because some patients continue to have optic
700
nerve deterioration despite proper IOP control. This indicates that the need for complementary
701
approaches for the treatment of glaucoma is higher than ever. Considering glaucoma as a
702
neurodegenerative disease, the development of a neuroprotective strategy may provide improved
703
therapeutic options for all glaucoma patients. However, until now, and even despite good laboratory
704
evidence, no neuroprotective agent is yet available for glaucoma treatment. This could very well be
705
explained by the narrow window of their neuroprotective actions. This review summarizes
706
accumulating experimental evidence that ROCK inhibitors not only promote RGC survival, but also
707
RGC axon regeneration and ocular blood flow. Much of the progress that was recently made
708
concerning the potential of ROCK inhibition for neuroprotection and axonal regeneration has been
709
obtained from several (animal) models of glaucoma. These findings increased our knowledge on the
710
beneficial effects by which ROCK inhibitors support cell survival and axonal regrowth, and also
711
unveiled some contributing signaling pathways. In order to reach improved survival of injured
712
neurons and inhibition of repulsive environmental signaling in glaucoma, combinatorial,
713
complementary pharmacotherapies that target multiple pathogenic processes/signaling pathways
28
714
might have a higher potential than those targeting a single molecule. Therefore, it is important that
715
drug discovery work at the in vitro, ex vivo and in vivo level is being complemented by studies that
716
aim to elucidate the downstream signaling pathways by which ROCK inhibition establishes its effects.
717
Overall, to establish neuroprotection and axonal regeneration as effective treatment modalities in
718
order to improve long-term outcomes, potent, preferably locally acting, ROCK inhibitors may be a
719
novel pharmacological approach for glaucoma.
720
721
Conflict of interest
722
All authors report to have no conflict of interest.
723
724
Acknowledgments
725
The authors are financially supported by the Hercules Grant [AKUL/09/038] and national Grants from
726
the Research Council of KU Leuven [KU Leuven BOF-OT/10/033; GOA/12/008] and the Flemish
727
Institute for the promotion of scientific research (IWT and FWO).
728
29
729
Figures
730
731
Figure 1. Hypothetical cascade of events leading to RGC degeneration in glaucoma.
732
Both mechanical and/or ischemic insults may cause mitochondrial dysfunction, thereby resulting in
733
oxidative stress and subsequent RGC apoptosis due to oxidative damage. Mitochondrial dysfunction
734
compromises RGC energy supply rendering RGCs to exist in a lower energetic state. Upon insult, glial
735
cells will become reactivated causing a gradual release of growth factors and other molecules, such
736
as TNF-α, glutamate, prostaglandins (PGs) and nitric oxide (NO). Initially, Müller glia will try to
737
maintain a physiological balance, but eventually, this process will become insufficient. A gradual
738
increase in the level of these potential harmful molecules will exist and a toxic environment will be
739
established. Because RGCs are already in a decreased energetic state, they are even more susceptible
740
to further injury and as a result, also these RGCs will be triggered to death via apoptosis.
741
742
30
743
744
745
Figure 2. Downstream targets of ROCK.
746
Activation of ROCK by GTP-bound RhoA leads to phosphorylation of several substrates regulating a
747
diverse range of cellular responses. Most downstream targets of ROCKs are important cytoskeletal
748
regulators. Hence, the Rho-ROCK pathway has critical functions in cytoskeletal based activities such
749
as, cell adhesion, morphology, motility and contraction. Several other downstream effectors are
750
highly expressed in the central nervous system (CNS) (dotted lines), and are regulators of diverse
751
neural processes, ranging from neuronal survival to axon outgrowth and guidance (GDP: guanosine
752
diphosphate; GTP: guanosine triphosphate; GAP: GTP-ase activating proteins; GEF: guanine
753
nucleotide-exchange factors; ROCK: Rho-associated coiled-coil protein kinase; MLC: myosin light
754
chain; MLCP: MLC phosphatase; LIMK: LIM kinase; ERM: ezrin/radixin/moesin; MAP: microtubule-
755
associated protein; CRMP-2: collapsin response mediator protein-2; GFAP: glial fibrillary acidic
756
protein; NFs: neurofilament; PTEN: phosphatase and tensin homolog).
757
758
759
31
760
761
Figure 3. Effect of ROCK inhibitors on neurite outgrowth in mouse retinal explants.
762
(A) The neurite outgrowth-stimulating effect of Y-27632 (10µM) and Y-39983 (10µM) was
763
demonstrated in retinal explants, harvested from P3 mouse pups and immunostained for β-tubulin
764
after 3 days in culture, via automatic analysis of the immunostained neurite outgrowth area.
765
Outgrowth in control explants was set at 100%. A combinatorial treatment with the growth factors,
766
BDNF (50 ng/ml) and CNTF (10 ng/ml), was used as a positive control. Both ROCK inhibitors, but
767
mainly Y-39983, significantly promoted neurite outgrowth. (B) Neurite outgrowth extension was
768
investigated by categorizing neurite outgrowth into 4 segments, generated by successive 100 µm
769
increments in radius from the explant body edge and thus resulting in three ring segments, and an
770
outer segment covering the remaining immunolabeled neurites. BDNF/CNTF treatment clearly
771
stimulated axon extension, yet, no significant neurite-extension-promoting effect could be observed
772
for any of the ROCK inhibitors. Data are shown as mean ± SEM (n ≥ 34 for every condition, from 4
773
independent experiments; ** p < 0.01, *** p < 0.001, two-way ANOVA with Bonferroni post hoc
774
test).
32
Table 1. Overview of observed axon outgrowth/regenerative effects of ROCK inhibitors in experimental glaucoma models
Drug
Optimal
Dosage form
Model
Animal
Effects
Reference
reduced repulsive neurite outgrowth
(Monnier et al.
dose
Y-27632
50µM
E6
retinal
explants chicken
2003)
(+CSPG)
Y-27632
10-100µM
adult retinal explants
cat
Y-39983: more neurite outgrowth
(Sagawa et al.
Y-39983
3 & 10µM
adult retinal explants
cat
similar effects on neurite length
2007)
increased amount glial processes
(Sagawa et al.
2007)
Y-39983
10µM
Gel
foam
+ ONC
cat
increased amount and length of reg. axons
(Sagawa et al.
2007)
injection at crush +
IV D0 (+7)
Y-27632
1.5mM
IV D0, 3, 6
ONC
rat
increased amount of reg. axons
(Lingor et al.
dimethylfasudil
4nM-40µM;
IV D0, 3, 6
ONC
rat
no effect on axonal outgrowth
2007)
(Lingor et al.
2007)
33
Y-27632
Y-27632
10µM
RGC-5 cells
10µM
P7-8 pRGCs
enhanced neurite outgrowth
(Lingor et al.
rat
enhanced neurite outgrowth
2008)
2.9mM
IV D0, 3, 6
peri. nerve graft
rat
no effect on axon number
2.9mM
IV D0, 3, 6
ONC
rat
increased amount reg. axons
adult pRGCs
rat
no effect on neurite outgrowth in absence or (Ahmed et al.
10µM
(+ myelin)
Y-27632
30 & 100µM
adult retinal explants
presence of myelin
cat
increased amount of neurites & glia, no effect (Ichikawa et al.
on process length
fasudil
10 & 30µM
adult retinal explants
cat
2009)
2008)
increased amount of glia, not neurites, no
effect on length
Y-27632
10 & 100µM
IV D0, 7 + gel foam
ONC
cat
increased amount and length of regenerating
fasudil
10-100µM
IV D0, 7 + gel foam
ONC
cat
axons
no regenerating axons
(Ichikawa et al.
2008)
(Ichikawa et al.
2008)
(Ichikawa et al.
2008)
34
Y-27632
10µM
RGC-5 cells
10µM
P7-8 pRGCs (+ CSPG)
10µM
adult
retinal
rat
explants rat
significantly increased neurite length
(Bermel et al.
enhanced neurite outgrowth
2009)
increased amount of outgrowing axons
after in vivo ONC
3.3µg
Y-39983
Y-27632
IV D0, 3
10µM
rat
trend towards more reg. axons
P7 pRGCs
rat
extended neurite outgrowth
(Tokushige
IV D0 + sponge
peri. nerve graft
rat
increased amount regenerating axons
al. 2011)
3mM
IV D0, 7
ONC
mouse
no effect on axonal outgrowth
(Pernet et al.
P7 pRGCs
rat
enhanced neurite outgrowth
2013)
ONC
rat
increased amount of reg. axons
(Koch
IV D-14
et
2014)
Abbreviations: D: day of injection; pRGCs: primary retinal ganglion cells; ONC: optic nerve crush; RGCL: retinal ganglion cell layer, E: embryonic day; P:
postnatal day; CSPG: chondroitin sulfate proteoglycan
35
et
10µM
AAV.ROCK2shRNA
ONC
al.
Table 2. Overview of observed neuroprotective effects of ROCK inhibitors in experimental glaucoma models
Drug
Optimal dose
Y-27632
10µM
Dosage form
Model
Animal
Effects
Reference
P7-8 pRGCs (-NT)
rat
increased cell survival
(Bermel et
al. 2009)
Y-27632
100nM
IV, D0
transient retinal ischemia
rat
reduced cell loss in the RGCL
(Hirata et
increased IPL thickness
al. 2008)
diminished leukocyte infiltration & endothelial
disarrangment
Y-27632
(Lingor et
rat
increased RGC survival
al. 2008)
ONT
rat
increased RGC survival
adult retinal explant
cat
no increased RGC survival
RGC-5 (-serum)
10µM
P7-8 pRGCs (-NTs)
2.9mM
Y-39983
increased RGC survival
10µM
IV, D0,3,6
10 & 30µM
(Sagawa et
al. 2007)
Fasudil
10 & 100µM
IV, D0
NMDA excitotoxicity
rat
reduced cell loss in the RGCL
(Kitaoka et
increased IPL thickness
al. 2004)
elevated Thy-1 mRNA levels
36
Fasudil
K-115
1mg/kg
1mg/kg
oral, daily
oral, daily
ONC
mouse
ONC
mouse
increased RGC survival
increased Thy-1 mRNA levels
(Yamamot
increased RGC survival
o
reduced oxidative stress
2014)
et
al.
(Yamamot
o
et
al.
2014)
dimethylfasudil
1µM
adult retinal explant
mouse
(-serum)
1, 10 & 100µM
IV D0
ONC
rat
reduced cell death
(Tura et al.
reduced macroglia reactivity
2009)
reduced cell death in the RGCL
reduced macro/microglia activity
dimethylfasudil
1-5µM
retinal
explant
(+ calf
hypoxia)
AAV.ROCK2shRNA
IV D-14
rat
ONC
reduced cell death
(Alt et al.
reduced macro/microglia activity
2013)
increased RGC survival
(Koch et al.
2014)
37
Abbreviations: D: day of injection; pRGCs: primary retinal ganglion cells; NT: neurotrophins; P: postnatal day; ONT: optic nerve transection;
ONC: optic nerve crush; RGCL: retinal ganglion cell layer; IPL: inner plexiform layer
38
References
Addicks, E. M., Quigley, H. A., Green, W. R. and Robin, A. L. (1983). "Histologic characteristics of
filtering blebs in glaucomatous eyes." Arch Ophthalmol 101(5): 795-798.
Agarwal, R., Gupta, S. K., Agarwal, P., Saxena, R. and Agrawal, S. S. (2009). "Current concepts in the
pathophysiology of glaucoma." Indian J Ophthalmol 57(4): 257-266.
Ahmed, Z., Berry, M. and Logan, A. (2009). "ROCK inhibition promotes adult retinal ganglion cell
neurite outgrowth only in the presence of growth promoting factors." Mol Cell Neurosci
42(2): 128-133.
Akhter, N., Nix, M., Abdul, Y., Singh, S. and Husain, S. (2013). "Delta-opioid receptors attenuate TNFalpha-induced MMP-2 secretion from human ONH astrocytes." Invest Ophthalmol Vis Sci
54(10): 6605-6611.
Alabed, Y. Z., Grados-Munro, E., Ferraro, G. B., Hsieh, S. H. and Fournier, A. E. (2006). "Neuronal
responses to myelin are mediated by rho kinase." J Neurochem 96(6): 1616-1625.
Alt, A., Hilgers, R. D., Tura, A., Nassar, K., Schneider, T., Hueber, A., Januschowski, K., Grisanti, S.,
Luke, J. and Luke, M. (2013). "The neuroprotective potential of Rho-kinase inhibition in
promoting cell survival and reducing reactive gliosis in response to hypoxia in isolated bovine
retina." Cell Physiol Biochem 32(1): 218-234.
Amano, M., Kaneko, T., Maeda, A., Nakayama, M., Ito, M., Yamauchi, T., Goto, H., Fukata, Y., Oshiro,
N., Shinohara, A., Iwamatsu, A. and Kaibuchi, K. (2003). "Identification of Tau and MAP2 as
novel substrates of Rho-kinase and myosin phosphatase." J Neurochem 87(3): 780-790.
Anastassiadis, T., Deacon, S. W., Devarajan, K., Ma, H. and Peterson, J. R. (2011). "Comprehensive
assay of kinase catalytic activity reveals features of kinase inhibitor selectivity." Nat
Biotechnol 29(11): 1039-1045.
Arimura, N., Inagaki, N., Chihara, K., Menager, C., Nakamura, N., Amano, M., Iwamatsu, A., Goshima,
Y. and Kaibuchi, K. (2000). "Phosphorylation of collapsin response mediator protein-2 by Rhokinase. Evidence for two separate signaling pathways for growth cone collapse." J Biol Chem
275(31): 23973-23980.
Arita, R., Hata, Y., Nakao, S., Kita, T., Miura, M., Kawahara, S., Zandi, S., Almulki, L., Tayyari, F.,
Shimokawa, H., Hafezi-Moghadam, A. and Ishibashi, T. (2009). "Rho kinase inhibition by
fasudil ameliorates diabetes-induced microvascular damage." Diabetes 58(1): 215-226.
Baltmr, A., Duggan, J., Nizari, S., Salt, T. E. and Cordeiro, M. F. (2010). "Neuroprotection in glaucoma Is there a future role?" Exp Eye Res 91(5): 554-566.
Bermel, C., Tonges, L., Planchamp, V., Gillardon, F., Weishaupt, J. H., Dietz, G. P., Bahr, M. and Lingor,
P. (2009). "Combined inhibition of Cdk5 and ROCK additively increase cell survival, but not
the regenerative response in regenerating retinal ganglion cells." Mol Cell Neurosci 42(4):
427-437.
Bishop, A. L. and Hall, A. (2000). "Rho GTPases and their effector proteins." Biochem J 348 Pt 2: 241255.
Bodor, N. and Buchwald, P. (2008). "Retrometabolic drug design: principles and recent
developments." Pure Appl Chem 80(8): 1669-1682.
Boland, M. V. and Quigley, H. A. (2007). "Risk factors and open-angle glaucoma: classification and
application." J Glaucoma 16(4): 406-418.
Boland, S., Defert, O., Alen, J., Bourin, A., Castermans, K., Kindt, N., Boumans, N., Panitti, L., Van de
Velde, S., Stalmans, I. and Leysen, D. (2013). "3-[2-(Aminomethyl)-5-[(pyridin-4yl)carbamoyl]phenyl] benzoates as soft ROCK inhibitors." Bioorg Med Chem Lett.
Bonomi, L., Marchini, G., Marraffa, M., Bernardi, P., Morbio, R. and Varotto, A. (2000). "Vascular risk
factors for primary open angle glaucoma: the Egna-Neumarkt Study." Ophthalmology 107(7):
1287-1293.
Borrajo, A., Rodriguez-Perez, A. I., Villar-Cheda, B., Guerra, M. J. and Labandeira-Garcia, J. L. (2014).
"Inhibition of the microglial response is essential for the neuroprotective effects of Rhokinase inhibitors on MPTP-induced dopaminergic cell death." Neuropharmacology 85: 1-8.
39
Bos, J. L., Rehmann, H. and Wittinghofer, A. (2007). "GEFs and GAPs: critical elements in the control
of small G proteins." Cell 129(5): 865-877.
Bowerman, M., Murray, L. M., Boyer, J. G., Anderson, C. L. and Kothary, R. (2012). "Fasudil improves
survival and promotes skeletal muscle development in a mouse model of spinal muscular
atrophy." BMC medicine 10: 24.
Broadway, D. C. and Drance, S. M. (1998). "Glaucoma and vasospasm." Br J Ophthalmol 82(8): 862870.
Bull, N. D. and Martin, K. R. (2007). "Optic nerve restoration: new perspectives." J Glaucoma 16(5):
506-511.
Buyens, T., Gaublomme, D., Van Hove, I., De Groef, L. and Moons, L. (2014). "Quantitative
assessment of neurite outgrowth in mouse retinal explants." Methods Mol Biol 1162: 57-71.
Cai, D., Qiu, J., Cao, Z., McAtee, M., Bregman, B. S. and Filbin, M. T. (2001). "Neuronal cyclic AMP
controls the developmental loss in ability of axons to regenerate." J Neurosci 21(13): 47314739.
Chang, E. E. and Goldberg, J. L. (2012b). "Glaucoma 2.0: neuroprotection, neuroregeneration,
neuroenhancement." Ophthalmology 119(5): 979-986.
Chauhan, B. C. and Drance, S. M. (1992). "The relationship between intraocular pressure and visual
field progression in glaucoma." Graefes Arch Clin Exp Ophthalmol 230(6): 521-526.
Chen, J., Runyan, S. A. and Robinson, M. R. (2011). "Novel ocular antihypertensive compounds in
clinical trials." Clin Ophthalmol 5: 667-677.
Cheng, C., Webber, C. A., Wang, J., Xu, Y., Martinez, J. A., Liu, W. Q., McDonald, D., Guo, G. F.,
Nguyen, M. D. and Zochodne, D. W. (2008). "Activated RHOA and peripheral axon
regeneration." Exp Neurol 212(2): 358-369.
Cheung, W., Guo, L. and Cordeiro, M. F. (2008). "Neuroprotection in glaucoma: drug-based
approaches." Optom Vis Sci 85(6): 406-416.
Chrysostomou, V., Rezania, F., Trounce, I. A. and Crowston, J. G. (2013). "Oxidative stress and
mitochondrial dysfunction in glaucoma." Curr Opin Pharmacol 13(1): 12-15.
Dahlmann-Noor, A. H., Vijay, S., Limb, G. A. and Khaw, P. T. (2010). "Strategies for optic nerve rescue
and regeneration in glaucoma and other optic neuropathies." Drug Discov Today 15(7-8):
287-299.
Danesh-Meyer, H. V. (2011). "Neuroprotection in glaucoma: recent and future directions." Curr Opin
Ophthalmol 22(2): 78-86.
De Groef, L., Van Hove, I., Dekeyster, E., Stalmans, I. and Moons, L. (2014). "MMPs in the neuroretina
and optic nerve: modulators of glaucoma pathogenesis and repair?" Invest Ophthalmol Vis
Sci 55(3): 1953-1964.
de Kater, A. W., Spurr-Michaud, S. J. and Gipson, I. K. (1990). "Localization of smooth muscle myosincontaining cells in the aqueous outflow pathway." Invest Ophthalmol Vis Sci 31(2): 347-353.
de Lima, S., Koriyama, Y., Kurimoto, T., Oliveira, J. T., Yin, Y., Li, Y., Gilbert, H. Y., Fagiolini, M.,
Martinez, A. M. and Benowitz, L. (2012). "Full-length axon regeneration in the adult mouse
optic nerve and partial recovery of simple visual behaviors." Proc Natl Acad Sci U S A 109(23):
9149-9154.
Deyts, C., Galan-Rodriguez, B., Martin, E., Bouveyron, N., Roze, E., Charvin, D., Caboche, J. and
Betuing, S. (2009). "Dopamine D2 receptor stimulation potentiates PolyQ-Huntingtin-induced
mouse striatal neuron dysfunctions via Rho/ROCK-II activation." PloS one 4(12): e8287.
Ding, J., Li, Q. Y., Wang, X., Sun, C. H., Lu, C. Z. and Xiao, B. G. (2010). "Fasudil protects hippocampal
neurons against hypoxia-reoxygenation injury by suppressing microglial inflammatory
responses in mice." J Neurochem 114(6): 1619-1629.
Ding, J., Yu, J. Z., Li, Q. Y., Wang, X., Lu, C. Z. and Xiao, B. G. (2009). "Rho kinase inhibitor Fasudil
induces neuroprotection and neurogenesis partially through astrocyte-derived G-CSF." Brain
Behav Immun 23(8): 1083-1088.
Doe, C., Bentley, R., Behm, D. J., Lafferty, R., Stavenger, R., Jung, D., Bamford, M., Panchal, T.,
Grygielko, E., Wright, L. L., Smith, G. K., Chen, Z., Webb, C., Khandekar, S., Yi, T., Kirkpatrick,
40
R., Dul, E., Jolivette, L., Marino, J. P., Jr., Willette, R., Lee, D. and Hu, E. (2007). "Novel Rho
kinase inhibitors with anti-inflammatory and vasodilatory activities." J Pharmacol Exp Ther
320(1): 89-98.
Dong, C. J., Guo, Y., Agey, P., Wheeler, L. and Hare, W. A. (2008). "Alpha2 adrenergic modulation of
NMDA receptor function as a major mechanism of RGC protection in experimental glaucoma
and retinal excitotoxicity." Invest Ophthalmol Vis Sci 49(10): 4515-4522.
Duffy, P., Schmandke, A., Sigworth, J., Narumiya, S., Cafferty, W. B. and Strittmatter, S. M. (2009).
"Rho-associated kinase II (ROCKII) limits axonal growth after trauma within the adult mouse
spinal cord." J Neurosci 29(48): 15266-15276.
Etienne-Manneville, S. and Hall, A. (2002). "Rho GTPases in cell biology." Nature 420(6916): 629-635.
Fechtner, R. D. and Weinreb, R. N. (1994). "Mechanisms of optic nerve damage in primary open angle
glaucoma." Surv Ophthalmol 39(1): 23-42.
Fehlings, M. G., Theodore, N., Harrop, J., Maurais, G., Kuntz, C., Shaffrey, C. I., Kwon, B. K., Chapman,
J., Yee, A., Tighe, A. and McKerracher, L. (2011). "A phase I/IIa clinical trial of a recombinant
Rho protein antagonist in acute spinal cord injury." J Neurotrauma 28(5): 787-796.
Fernandes, L. B., Henry, P. J. and Goldie, R. G. (2007). "Rho kinase as a therapeutic target in the
treatment of asthma and chronic obstructive pulmonary disease." Ther Adv Respir Dis 1(1):
25-33.
Flammer, J. (1994). "The vascular concept of glaucoma." Surv Ophthalmol 38 Suppl: S3-6.
Flammer, J. and Mozaffarieh, M. (2007). "What is the present pathogenetic concept of glaucomatous
optic neuropathy?" Surv Ophthalmol 52 Suppl 2: S162-173.
Flammer, J., Orgül, S., Costa, V. P., Orzalesi, N., Krieglstein, G. K., Serra, L. M., Renard, J.-P. and
Stefánsson, E. (2002a). "The impact of ocular blood flow in glaucoma." Prog Retin Eye Res
21(4): 359-393.
Flammer, J., Orgul, S., Costa, V. P., Orzalesi, N., Krieglstein, G. K., Serra, L. M., Renard, J. P. and
Stefansson, E. (2002b). "The impact of ocular blood flow in glaucoma." Prog Retin Eye Res
21(4): 359-393.
Fleischman, D. and Allingham, R. R. (2013). "The role of cerebrospinal fluid pressure in glaucoma and
other ophthalmic diseases: A review." Saudi J Ophthalmol 27(2): 97-106.
Foster, P. J., Buhrmann, R., Quigley, H. A. and Johnson, G. J. (2002). "The definition and classification
of glaucoma in prevalence surveys." Br J Ophthalmol 86(2): 238-242.
Fournier, A. E., Takizawa, B. T. and Strittmatter, S. M. (2003). "Rho kinase inhibition enhances axonal
regeneration in the injured CNS." J Neurosci 23(4): 1416-1423.
Friedman, D. S., Wolfs, R. C., O'Colmain, B. J., Klein, B. E., Taylor, H. R., West, S., Leske, M. C.,
Mitchell, P., Congdon, N. and Kempen, J. (2004). "Prevalence of open-angle glaucoma among
adults in the United States." Arch Ophthalmol 122(4): 532-538.
Froger, N., Cadetti, L., Lorach, H., Martins, J., Bemelmans, A. P., Dubus, E., Degardin, J., Pain, D.,
Forster, V., Chicaud, L., Ivkovic, I., Simonutti, M., Fouquet, S., Jammoul, F., Leveillard, T.,
Benosman, R., Sahel, J. A. and Picaud, S. (2012). "Taurine provides neuroprotection against
retinal ganglion cell degeneration." PLoS One 7(10): e42017.
Fujita, Y. and Yamashita, T. (2014). "Axon growth inhibition by RhoA/ROCK in the central nervous
system." Front Neurosci 8: 338.
Fukunaga, T., Ikesugi, K., Nishio, M., Sugimoto, M., Sasoh, M., Hidaka, H. and Uji, Y. (2009). "The
effect of the Rho-associated protein kinase inhibitor, HA-1077, in the rabbit ocular
hypertension model induced by water loading." Curr Eye Res 34(1): 42-47.
Gallo, G. (2006). "RhoA-kinase coordinates F-actin organization and myosin II activity during
semaphorin-3A-induced axon retraction." J Cell Sci 119(Pt 16): 3413-3423.
Gao, C., Huang, L., Long, Y., Zheng, J., Yang, J., Pu, S. and Xie, L. (2013). "Y-39983, a selective Rhokinase inhibitor, attenuates experimental autoimmune encephalomyelitis via inhibition of
demyelination." Neuroimmunomodulation 20(6): 334-340.
Gaublomme, D., Buyens, T. and Moons, L. (2013). "Automated analysis of neurite outgrowth in
mouse retinal explants." J Biomol Screen 18(5): 534-543.
41
Goel, M., Picciani, R. G., Lee, R. K. and Bhattacharya, S. K. (2010). "Aqueous humor dynamics: a
review." Open Ophthalmol J 4: 52-59.
Goldhagen, B., Proia, A. D., Epstein, D. L. and Rao, P. V. (2012). "Elevated levels of RhoA in the optic
nerve head of human eyes with glaucoma." J Glaucoma 21(8): 530-538.
Gopalakrishnan, S. M., Teusch, N., Imhof, C., Bakker, M. H., Schurdak, M., Burns, D. J. and Warrior, U.
(2008). "Role of Rho kinase pathway in chondroitin sulfate proteoglycan-mediated inhibition
of neurite outgrowth in PC12 cells." J Neurosci Res 86(10): 2214-2226.
Gungabissoon, R. A. and Bamburg, J. R. (2003). "Regulation of growth cone actin dynamics by
ADF/cofilin." J Histochem Cytochem 51(4): 411-420.
Gunther, R., Saal, K. A., Suhr, M., Scheer, D., Koch, J. C., Bahr, M., Lingor, P. and Tonges, L. (2014).
"The rho kinase inhibitor Y-27632 improves motor performance in male SOD1(G93A) mice."
Front Neurosci 8: 304.
Hasegawa, Y., Fujitani, M., Hata, K., Tohyama, M., Yamagishi, S. and Yamashita, T. (2004). "Promotion
of axon regeneration by myelin-associated glycoprotein and Nogo through divergent signals
downstream of Gi/G." J Neurosci 24(30): 6826-6832.
Hashimoto, R., Nakamura, Y., Goto, H., Wada, Y., Sakoda, S., Kaibuchi, K., Inagaki, M. and Takeda, M.
(1998). "Domain- and site-specific phosphorylation of bovine NF-L by Rho-associated kinase."
Biochem Biophys Res Commun 245(2): 407-411.
Hayreh, S. S. (1999). "The role of age and cardiovascular disease in glaucomatous optic neuropathy."
Surv Ophthalmol 43 Suppl 1: S27-42.
Hayreh, S. S., Zimmerman, M. B., Podhajsky, P. and Alward, W. L. (1994). "Nocturnal arterial
hypotension and its role in optic nerve head and ocular ischemic disorders." Am J
Ophthalmol 117(5): 603-624.
He, Y., Xu, H., Liang, L., Zhan, Z., Yang, X., Yu, X., Ye, Y. and Sun, L. (2008). "Antiinflammatory effect of
Rho kinase blockade via inhibition of NF-kappaB activation in rheumatoid arthritis." Arthritis
Rheum 58(11): 3366-3376.
Hernandez, M. R. (2000). "The optic nerve head in glaucoma: role of astrocytes in tissue remodeling."
Prog Retin Eye Res 19(3): 297-321.
Herskowitz, J. H., Feng, Y., Mattheyses, A. L., Hales, C. M., Higginbotham, L. A., Duong, D. M.,
Montine, T. J., Troncoso, J. C., Thambisetty, M., Seyfried, N. T., Levey, A. I. and Lah, J. J.
(2013). "Pharmacologic inhibition of ROCK2 suppresses amyloid-beta production in an
Alzheimer's disease mouse model." J Neurosci 33(49): 19086-19098.
Hiraga, A., Kuwabara, S., Doya, H., Kanai, K., Fujitani, M., Taniguchi, J., Arai, K., Mori, M., Hattori, T.
and Yamashita, T. (2006). "Rho-kinase inhibition enhances axonal regeneration after
peripheral nerve injury." J Peripher Nerv Syst 11(3): 217-224.
Hirata, A., Inatani, M., Inomata, Y., Yonemura, N., Kawaji, T., Honjo, M. and Tanihara, H. (2008). "Y27632, a Rho-associated protein kinase inhibitor, attenuates neuronal cell death after
transient retinal ischemia." Graefes Arch Clin Exp Ophthalmol 246(1): 51-59.
Honjo, M., Inatani, M., Kido, N., Sawamura, T., Yue, B. Y., Honda, Y. and Tanihara, H. (2001a). "Effects
of protein kinase inhibitor, HA1077, on intraocular pressure and outflow facility in rabbit
eyes." Arch Ophthalmol 119(8): 1171-1178.
Honjo, M., Tanihara, H., Inatani, M., Kido, N., Sawamura, T., Yue, B. Y., Narumiya, S. and Honda, Y.
(2001b). "Effects of rho-associated protein kinase inhibitor Y-27632 on intraocular pressure
and outflow facility." Invest Ophthalmol Vis Sci 42(1): 137-144.
Honjo, M., Tanihara, H., Kameda, T., Kawaji, T., Yoshimura, N. and Araie, M. (2007). "Potential role of
Rho-associated protein kinase inhibitor Y-27632 in glaucoma filtration surgery." Invest
Ophthalmol Vis Sci 48(12): 5549-5557.
Ichikawa, M., Yoshida, J., Saito, K., Sagawa, H., Tokita, Y. and Watanabe, M. (2008). "Differential
effects of two ROCK inhibitors, Fasudil and Y-27632, on optic nerve regeneration in adult
cats." Brain Res 1201: 23-33.
Isobe, T., Mizuno, K., Kaneko, Y., Ohta, M., Koide, T. and Tanabe, S. (2014). "Effects of k-115, a rhokinase inhibitor, on aqueous humor dynamics in rabbits." Curr Eye Res 39(8): 813-822.
42
Itoh, K., Yoshioka, K., Akedo, H., Uehata, M., Ishizaki, T. and Narumiya, S. (1999). "An essential part
for Rho-associated kinase in the transcellular invasion of tumor cells." Nat Med 5(2): 221225.
Ji, J. Z., Elyaman, W., Yip, H. K., Lee, V. W., Yick, L. W., Hugon, J. and So, K. F. (2004a). "CNTF promotes
survival of retinal ganglion cells after induction of ocular hypertension in rats: the possible
involvement of STAT3 pathway." Eur J Neurosci 19(2): 265-272.
Ji, J. Z., Elyaman, W., Yip, H. K., Lee, V. W., Yick, L. W., Hugon, J. and So, K. F. (2004b). "CNTF promotes
survival of retinal ganglion cells after induction of ocular hypertension in rats: the possible
involvement of STAT3 pathway." Eur J Neurosci 19(2): 265-272.
Johnson, E. C. and Morrison, J. C. (2009). "Friend or foe? Resolving the impact of glial responses in
glaucoma." J Glaucoma 18(5): 341-353.
Johnson, T. V., Bull, N. D. and Martin, K. R. (2011). "Neurotrophic factor delivery as a protective
treatment for glaucoma." Exp Eye Res 93(2): 196-203.
Kaneko-Kawano, T., Takasu, F., Naoki, H., Sakumura, Y., Ishii, S., Ueba, T., Eiyama, A., Okada, A.,
Kawano, Y. and Suzuki, K. (2012). "Dynamic regulation of myosin light chain phosphorylation
by Rho-kinase." PLoS One 7(6): e39269.
Kaplan, D. R. and Miller, F. D. (2003). "Axon growth inhibition: signals from the p75 neurotrophin
receptor." Nat Neurosci 6(5): 435-436.
Kass, M. A., Heuer, D. K., Higginbotham, E. J., Johnson, C. A., Keltner, J. L., Miller, J. P., Parrish, R. K.,
2nd, Wilson, M. R. and Gordon, M. O. (2002). "The Ocular Hypertension Treatment Study: a
randomized trial determines that topical ocular hypotensive medication delays or prevents
the onset of primary open-angle glaucoma." Arch Ophthalmol 120(6): 701-713; discussion
829-730.
Kast, R., Schirok, H., Figueroa-Perez, S., Mittendorf, J., Gnoth, M. J., Apeler, H., Lenz, J., Franz, J. K.,
Knorr, A., Hutter, J., Lobell, M., Zimmermann, K., Munter, K., Augstein, K. H., Ehmke, H. and
Stasch, J. P. (2007). "Cardiovascular effects of a novel potent and highly selective azaindolebased inhibitor of Rho-kinase." Br J Pharmacol 152(7): 1070-1080.
Kirwan, R. P., Crean, J. K., Fenerty, C. H., Clark, A. F. and O'Brien, C. J. (2004). "Effect of cyclical
mechanical stretch and exogenous transforming growth factor-beta1 on matrix
metalloproteinase-2 activity in lamina cribrosa cells from the human optic nerve head." J
Glaucoma 13(4): 327-334.
Kitaoka, Y., Kumai, T., Lam, T. T., Kuribayashi, K., Isenoumi, K., Munemasa, Y., Motoki, M., Kobayashi,
S. and Ueno, S. (2004). "Involvement of RhoA and possible neuroprotective effect of fasudil,
a Rho kinase inhibitor, in NMDA-induced neurotoxicity in the rat retina." Brain Res 1018(1):
111-118.
Koch, J. C., Tonges, L., Barski, E., Michel, U., Bahr, M. and Lingor, P. (2014). "ROCK2 is a major
regulator of axonal degeneration, neuronal death and axonal regeneration in the CNS." Cell
Death Dis 5: e1225.
Koumura, A., Hamanaka, J., Kawasaki, K., Tsuruma, K., Shimazawa, M., Hozumi, I., Inuzuka, T. and
Hara, H. (2011). "Fasudil and ozagrel in combination show neuroprotective effects on
cerebral infarction after murine middle cerebral artery occlusion." J Pharmacol Exp Ther
338(1): 337-344.
Krishnamoorthy, R. R., Clark, A. F., Daudt, D., Vishwanatha, J. K. and Yorio, T. (2013). "A forensic path
to RGC-5 cell line identification: lessons learned." Invest Ophthalmol Vis Sci 54(8): 5712-5719.
Krupin, T., Liebmann, J. M., Greenfield, D. S., Ritch, R. and Gardiner, S. (2011). "A randomized trial of
brimonidine versus timolol in preserving visual function: results from the Low-Pressure
Glaucoma Treatment Study." Am J Ophthalmol 151(4): 671-681.
Kubo, T., Yamaguchi, A., Iwata, N. and Yamashita, T. (2008). "The therapeutic effects of Rho-ROCK
inhibitors on CNS disorders." Ther Clin Risk Manag 4(3): 605-615.
Kume, H. (2008). "RhoA/Rho-kinase as a therapeutic target in asthma." Curr Med Chem 15(27): 28762885.
43
Labandeira-Garcia, J. L., Rodriguez-Perez, A. I., Villar-Cheda, B., Borrajo, A., Dominguez-Meijide, A.
and Guerra, M. J. (2014). "Rho Kinase and Dopaminergic Degeneration: A Promising
Therapeutic Target for Parkinson's Disease." Neuroscientist
Lai, R. K., Chun, T., Hasson, D., Lee, S., Mehrbod, F. and Wheeler, L. (2002). "Alpha-2 adrenoceptor
agonist protects retinal function after acute retinal ischemic injury in the rat." Vis Neurosci
19(2): 175-185.
Lee, D. A. and Higginbotham, E. J. (2005). "Glaucoma and its treatment: a review." Am J Health Syst
Pharm 62(7): 691-699.
Lehwalder, D., Jeffrey, P. L. and Unsicker, K. (1989). "Survival of purified embryonic chick retinal
ganglion cells in the presence of neurotrophic factors." J Neurosci Res 24(2): 329-337.
Leibinger, M., Muller, A., Andreadaki, A., Hauk, T. G., Kirsch, M. and Fischer, D. (2009).
"Neuroprotective and axon growth-promoting effects following inflammatory stimulation on
mature retinal ganglion cells in mice depend on ciliary neurotrophic factor and leukemia
inhibitory factor." J Neurosci 29(45): 14334-14341.
Leung, T., Chen, X. Q., Manser, E. and Lim, L. (1996). "The p160 RhoA-binding kinase ROK alpha is a
member of a kinase family and is involved in the reorganization of the cytoskeleton." Mol
Cell Biol 16(10): 5313-5327.
Li, M., Huang, Y., Ma, A. A., Lin, E. and Diamond, M. I. (2009). "Y-27632 improves rotarod
performance and reduces huntingtin levels in R6/2 mice." Neurobiol Dis 36(3): 413-420.
Li, Y. H., Yu, J. Z., Liu, C. Y., Zhang, H., Zhang, H. F., Yang, W. F., Li, J. L., Feng, Q. J., Feng, L., Zhang, G.
X., Xiao, B. G. and Ma, C. G. (2014a). "Intranasal delivery of FSD-C10, a novel Rho kinase
inhibitor, exhibits therapeutic potential in experimental autoimmune encephalomyelitis."
Immunology 143(2):219-229.
Li, Z., Dong, X., Wang, Z., Liu, W., Deng, N., Ding, Y., Tang, L., Hla, T., Zeng, R., Li, L. and Wu, D. (2005).
"Regulation of PTEN by Rho small GTPases." Nat Cell Biol 7(4): 399-404.
Liao, J. K., Seto, M. and Noma, K. (2007). "Rho kinase (ROCK) inhibitors." J Cardiovasc Pharmacol
50(1): 17-24.
Lingor, P., Teusch, N., Schwarz, K., Mueller, R., Mack, H., Bahr, M. and Mueller, B. K. (2007).
"Inhibition of Rho kinase (ROCK) increases neurite outgrowth on chondroitin sulphate
proteoglycan in vitro and axonal regeneration in the adult optic nerve in vivo." J Neurochem
103(1): 181-189.
Lingor, P., Tonges, L., Pieper, N., Bermel, C., Barski, E., Planchamp, V. and Bahr, M. (2008). "ROCK
inhibition and CNTF interact on intrinsic signalling pathways and differentially regulate
survival and regeneration in retinal ganglion cells." Brain 131(Pt 1): 250-263.
Llobet, A., Gasull, X. and Gual, A. (2003). "Understanding trabecular meshwork physiology: a key to
the control of intraocular pressure?" News Physiol Sci 18: 205-209.
LoGrasso, P. V. and Feng, Y. (2009). "Rho kinase (ROCK) inhibitors and their application to
inflammatory disorders." Curr Top Med Chem 9(8): 704-723.
Lorber, B., Berry, M., Douglas, M. R., Nakazawa, T. and Logan, A. (2009). "Activated retinal glia
promote neurite outgrowth of retinal ganglion cells via apolipoprotein E." J Neurosci Res
87(12): 2645-2652.
Lorber, B., Guidi, A., Fawcett, J. W. and Martin, K. R. (2012). "Activated retinal glia mediated axon
regeneration in experimental glaucoma." Neurobiol Dis 45(1): 243-252.
Lu, Z., Overby, D. R., Scott, P. A., Freddo, T. F. and Gong, H. (2008). "The mechanism of increasing
outflow facility by rho-kinase inhibition with Y-27632 in bovine eyes." Exp Eye Res 86(2): 271281.
Luo, X., Salgueiro, Y., Beckerman, S. R., Lemmon, V. P., Tsoulfas, P. and Park, K. K. (2013). "Threedimensional evaluation of retinal ganglion cell axon regeneration and pathfinding in whole
mouse tissue after injury." Exp Neurol 247: 653-662.
Maier, K., Rau, C. R., Storch, M. K., Sattler, M. B., Demmer, I., Weissert, R., Taheri, N., Kuhnert, A. V.,
Bahr, M. and Diem, R. (2004). "Ciliary neurotrophic factor protects retinal ganglion cells from
44
secondary cell death during acute autoimmune optic neuritis in rats." Brain Pathol 14(4):
378-387.
Mandell, K. J., Kudelka, R. M. and Wirostko, B. (2011). "Rho kinase inhibitors for treatment of
glaucoma." Expert Rev Ophthalmol 6(6): 611-622.
Martinez-Bello, C., Chauhan, B. C., Nicolela, M. T., McCormick, T. A. and LeBlanc, R. P. (2000).
"Intraocular pressure and progression of glaucomatous visual field loss." Am J Ophthalmol
129(3): 302-308.
Matsui, T., Amano, M., Yamamoto, T., Chihara, K., Nakafuku, M., Ito, M., Nakano, T., Okawa, K.,
Iwamatsu, A. and Kaibuchi, K. (1996). "Rho-associated kinase, a novel serine/threonine
kinase, as a putative target for small GTP binding protein Rho." EMBO J 15(9): 2208-2216.
Matsuoka, Y., Li, X. and Bennett, V. (2000). "Adducin: structure, function and regulation." Cell Mol
Life Sci 57(6): 884-895.
McKeon, R. J., Schreiber, R. C., Rudge, J. S. and Silver, J. (1991). "Reduction of neurite outgrowth in a
model of glial scarring following CNS injury is correlated with the expression of inhibitory
molecules on reactive astrocytes." J Neurosci 11(11): 3398-3411.
McKerracher, L. and Winton, M. J. (2002). "Nogo on the go." Neuron 36(3): 345-348.
Mey, J. and Thanos, S. (1993). "Intravitreal injections of neurotrophic factors support the survival of
axotomized retinal ganglion cells in adult rats in vivo." Brain Res 602(2): 304-317.
Meyer-ter-Vehn, T., Sieprath, S., Katzenberger, B., Gebhardt, S., Grehn, F. and Schlunck, G. (2006).
"Contractility as a prerequisite for TGF-beta-induced myofibroblast transdifferentiation in
human tenon fibroblasts." Invest Ophthalmol Vis Sci 47(11): 4895-4904.
Mimura, F., Yamagishi, S., Arimura, N., Fujitani, M., Kubo, T., Kaibuchi, K. and Yamashita, T. (2006).
"Myelin-associated glycoprotein inhibits microtubule assembly by a Rho-kinase-dependent
mechanism." J Biol Chem 281(23): 15970-15979.
Mitchell, P., Hourihan, F., Sandbach, J. and Wang, J. J. (1999). "The relationship between glaucoma
and myopia: the Blue Mountains Eye Study." Ophthalmology 106(10): 2010-2015.
Mitchell, P., Smith, W., Chey, T. and Healey, P. R. (1997). "Open-angle glaucoma and diabetes: the
Blue Mountains eye study, Australia." Ophthalmology 104(4): 712-718.
Monnier, P. P., Sierra, A., Schwab, J. M., Henke-Fahle, S. and Mueller, B. K. (2003). "The Rho/ROCK
pathway mediates neurite growth-inhibitory activity associated with the chondroitin sulfate
proteoglycans of the CNS glial scar." Mol Cell Neurosci 22(3): 319-330.
Morgan, J. E. (2004). "Circulation and axonal transport in the optic nerve." Eye (Lond) 18(11): 10891095.
Mueller, B. K., Mack, H. and Teusch, N. (2005). "Rho kinase, a promising drug target for neurological
disorders." Nat Rev Drug Discov 4(5): 387-398.
Muller, A., Hauk, T. G. and Fischer, D. (2007). "Astrocyte-derived CNTF switches mature RGCs to a
regenerative state following inflammatory stimulation." Brain 130(Pt 12): 3308-3320.
Murray, A. J. (2014). "Axon regeneration: what needs to be overcome?" Methods Mol Biol 1162: 314.
Nakagawa, O., Fujisawa, K., Ishizaki, T., Saito, Y., Nakao, K. and Narumiya, S. (1996). "ROCK-I and
ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase
in mice." FEBS Lett 392(2): 189-193.
Nakajima, E., Nakajima, T., Minagawa, Y., Shearer, T. R. and Azuma, M. (2005). "Contribution of ROCK
in contraction of trabecular meshwork: proposed mechanism for regulating aqueous outflow
in monkey and human eyes." J Pharm Sci 94(4): 701-708.
Nakazawa, T., Watabe, A. M., Tezuka, T., Yoshida, Y., Yokoyama, K., Umemori, H., Inoue, A., Okabe,
S., Manabe, T. and Yamamoto, T. (2003). "p250GAP, a novel brain-enriched GTPaseactivating protein for Rho family GTPases, is involved in the N-methyl-d-aspartate receptor
signaling." Mol Biol Cell 14(7): 2921-2934.
Ng, D. C., Lin, B. H., Lim, C. P., Huang, G., Zhang, T., Poli, V. and Cao, X. (2006). "Stat3 regulates
microtubules by antagonizing the depolymerization activity of stathmin." J Cell Biol 172(2):
245-257.
45
Nickells, R. W. (2007). "From ocular hypertension to ganglion cell death: a theoretical sequence of
events leading to glaucoma." Can J Ophthalmol 42(2): 278-287.
Nishio, M., Fukunaga, T., Sugimoto, M., Ikesugi, K., Sumi, K., Hidaka, H. and Uji, Y. (2009). "The effect
of the H-1152P, a potent Rho-associated coiled coil-formed protein kinase inhibitor, in rabbit
normal and ocular hypertensive eyes." Curr Eye Res 34(4): 282-286.
Norenberg, W., Hofmann, F., Illes, P., Aktories, K. and Meyer, D. K. (1999). "Rundown of
somatodendritic N-methyl-D-aspartate (NMDA) receptor channels in rat hippocampal
neurones: evidence for a role of the small GTPase RhoA." Br J Pharmacol 127(5): 1060-1063.
Ohashi, K., Nagata, K., Maekawa, M., Ishizaki, T., Narumiya, S. and Mizuno, K. (2000). "Rho-associated
kinase ROCK activates LIM-kinase 1 by phosphorylation at threonine 508 within the
activation loop." J Biol Chem 275(5): 3577-3582.
Osborne, N. N. (2009). "Recent clinical findings with memantine should not mean that the idea of
neuroprotection in glaucoma is abandoned." Acta Ophthalmol 87(4): 450-454.
Osborne, N. N., Melena, J., Chidlow, G. and Wood, J. P. (2001). "A hypothesis to explain ganglion cell
death caused by vascular insults at the optic nerve head: possible implication for the
treatment of glaucoma." Br J Ophthalmol 85(10): 1252-1259.
Park, K., Luo, J. M., Hisheh, S., Harvey, A. R. and Cui, Q. (2004). "Cellular mechanisms associated with
spontaneous and ciliary neurotrophic factor-cAMP-induced survival and axonal regeneration
of adult retinal ganglion cells." J Neurosci 24(48): 10806-10815.
Pasterkamp, R. J. and Verhaagen, J. (2006). "Semaphorins in axon regeneration: developmental
guidance molecules gone wrong?" Philos Trans R Soc Lond B Biol Sci 361(1473): 1499-1511.
Pernet, V., Joly, S., Jordi, N., Dalkara, D., Guzik-Kornacka, A., Flannery, J. G. and Schwab, M. E. (2013).
"Misguidance and modulation of axonal regeneration by Stat3 and Rho/ROCK signaling in the
transparent optic nerve." Cell Death Dis 4: e734.
Pernet, V. and Schwab, M. E. (2014). "Lost in the jungle: new hurdles for optic nerve axon
regeneration." Trends Neurosci 37(7): 381-387.
Quigley, H. A. (2011). "Glaucoma." Lancet 377(9774): 1367-1377.
Quigley, H. A. and Broman, A. T. (2006). "The number of people with glaucoma worldwide in 2010
and 2020." Br J Ophthalmol 90(3): 262-267.
Quigley, H. A., McKinnon, S. J., Zack, D. J., Pease, M. E., Kerrigan-Baumrind, L. A., Kerrigan, D. F. and
Mitchell, R. S. (2000). "Retrograde axonal transport of BDNF in retinal ganglion cells is
blocked by acute IOP elevation in rats." Invest Ophthalmol Vis Sci 41(11): 3460-3466.
Quill, B., Docherty, N. G., Clark, A. F. and O'Brien, C. J. (2011). "The effect of graded cyclic stretching
on extracellular matrix-related gene expression profiles in cultured primary human lamina
cribrosa cells." Invest Ophthalmol Vis Sci 52(3): 1908-1915.
Racette, L., Wilson, M. R., Zangwill, L. M., Weinreb, R. N. and Sample, P. A. (2003). "Primary openangle glaucoma in blacks: a review." Surv Ophthalmol 48(3): 295-313.
Rao, P. V., Deng, P. F., Kumar, J. and Epstein, D. L. (2001). "Modulation of aqueous humor outflow
facility by the Rho kinase-specific inhibitor Y-27632." Invest Ophthalmol Vis Sci 42(5): 10291037.
Resnikoff, S., Pascolini, D., Mariotti, S. P. and Pokharel, G. P. (2008). "Global magnitude of visual
impairment caused by uncorrected refractive errors in 2004." Bull World Health Organ 86(1):
63-70.
Riento, K., Guasch, R. M., Garg, R., Jin, B. and Ridley, A. J. (2003). "RhoE binds to ROCK I and inhibits
downstream signaling." Mol Cell Biol 23(12): 4219-4229.
Riento, K. and Ridley, A. J. (2003). "Rocks: multifunctional kinases in cell behaviour." Nat Rev Mol Cell
Biol 4(6): 446-456.
Rikitake, Y., Kim, H. H., Huang, Z., Seto, M., Yano, K., Asano, T., Moskowitz, M. A. and Liao, J. K.
(2005). "Inhibition of Rho kinase (ROCK) leads to increased cerebral blood flow and stroke
protection." Stroke 36(10): 2251-2257.
46
Rudnicka, A. R., Mt-Isa, S., Owen, C. G., Cook, D. G. and Ashby, D. (2006). "Variations in primary openangle glaucoma prevalence by age, gender, and race: a Bayesian meta-analysis." Invest
Ophthalmol Vis Sci 47(10): 4254-4261.
Sagawa, H., Terasaki, H., Nakamura, M., Ichikawa, M., Yata, T., Tokita, Y. and Watanabe, M. (2007).
"A novel ROCK inhibitor, Y-39983, promotes regeneration of crushed axons of retinal
ganglion cells into the optic nerve of adult cats." Exp Neurol 205(1): 230-240.
Satoh, S., Utsunomiya, T., Tsurui, K., Kobayashi, T., Ikegaki, I., Sasaki, Y. and Asano, T. (2001).
"Pharmacological profile of hydroxy fasudil as a selective rho kinase inhibitor on ischemic
brain damage." Life Sci 69(12): 1441-1453.
Schmandke, A. and Strittmatter, S. M. (2007). "ROCK and Rho: biochemistry and neuronal functions
of Rho-associated protein kinases." Neuroscientist 13(5): 454-469.
Selvaraj, B. T., Frank, N., Bender, F. L., Asan, E. and Sendtner, M. (2012). "Local axonal function of
STAT3 rescues axon degeneration in the pmn model of motoneuron disease." J Cell Biol
199(3): 437-451.
Shaw, R. J., Henry, M., Solomon, F. and Jacks, T. (1998). "RhoA-dependent phosphorylation and
relocalization of ERM proteins into apical membrane/actin protrusions in fibroblasts." Mol
Biol Cell 9(2): 403-419.
Shi, J. and Wei, L. (2013). "Rho kinases in cardiovascular physiology and pathophysiology: the effect
of fasudil." J Cardiovasc Pharmacol 62(4): 341-354.
Shi, J., Wu, X., Surma, M., Vemula, S., Zhang, L., Yang, Y., Kapur, R. and Wei, L. (2013). "Distinct roles
for ROCK1 and ROCK2 in the regulation of cell detachment." Cell Death Dis 4: e483.
Sigal, I. A. and Ethier, C. R. (2009). "Biomechanics of the optic nerve head." Exp Eye Res 88(4): 799807.
Silver, J. and Miller, J. H. (2004). "Regeneration beyond the glial scar." Nat Rev Neurosci 5(2): 146156.
Sippl, C. and Tamm, E. R. (2014). "What is the nature of the RGC-5 cell line?" Adv Exp Med Biol 801:
145-154.
Song, Y., Chen, X., Wang, L. Y., Gao, W. and Zhu, M. J. (2013). "Rho kinase inhibitor fasudil protects
against beta-amyloid-induced hippocampal neurodegeneration in rats." CNS Neurosci Ther
19(8): 603-610.
Stroman, G. A., Stewart, W. C., Golnik, K. C., Cure, J. K. and Olinger, R. E. (1995). "Magnetic resonance
imaging in patients with low-tension glaucoma." Arch Ophthalmol 113(2): 168-172.
Sugiyama, T., Shibata, M., Kajiura, S., Okuno, T., Tonari, M., Oku, H. and Ikeda, T. (2011). "Effects of
fasudil, a Rho-associated protein kinase inhibitor, on optic nerve head blood flow in rabbits."
Invest Ophthalmol Vis Sci 52(1): 64-69.
Sumi, T., Matsumoto, K. and Nakamura, T. (2001). "Specific activation of LIM kinase 2 via
phosphorylation of threonine 505 by ROCK, a Rho-dependent protein kinase." J Biol Chem
276(1): 670-676.
Sung, J. K., Miao, L., Calvert, J. W., Huang, L., Louis Harkey, H. and Zhang, J. H. (2003). "A possible role
of RhoA/Rho-kinase in experimental spinal cord injury in rat." Brain Res 959(1): 29-38.
Suzuki, Y., Shibuya, M., Satoh, S.-i., Sugimoto, Y. and Takakura, K. (2007). "A postmarketing
surveillance study of fasudil treatment after aneurysmal subarachnoid hemorrhage." Surg
Neurol 68(2): 126-131.
Tanihara, H., Inatani, M., Honjo, M., Tokushige, H., Azuma, J. and Araie, M. (2008). "Intraocular
pressure-lowering effects and safety of topical administration of a selective ROCK inhibitor,
SNJ-1656, in healthy volunteers." Arch Ophthalmol 126(3): 309-315.
Thanos, S., Naskar, R. and Heiduschka, P. (1997). "Regenerating ganglion cell axons in the adult rat
establish retinofugal topography and restore visual function." Exp Brain Res 114(3): 483-491.
Toft-Kehler, A. K., Skytt, D. M., Poulsen, K. A., Braendstrup, C. T., Gegelashvili, G., Waagepetersen, H.
and Kolko, M. (2014). "Limited energy supply in Muller cells alters glutamate uptake."
Neurochem Res 39(5): 941-949.
47
Tokushige, H., Inatani, M., Nemoto, S., Sakaki, H., Katayama, K., Uehata, M. and Tanihara, H. (2007).
"Effects of topical administration of y-39983, a selective rho-associated protein kinase
inhibitor, on ocular tissues in rabbits and monkeys." Invest Ophthalmol Vis Sci 48(7): 32163222.
Tokushige, H., Waki, M., Takayama, Y. and Tanihara, H. (2011). "Effects of Y-39983, a selective Rhoassociated protein kinase inhibitor, on blood flow in optic nerve head in rabbits and axonal
regeneration of retinal ganglion cells in rats." Curr Eye Res 36(10): 964-970.
Tonges, L., Frank, T., Tatenhorst, L., Saal, K. A., Koch, J. C., Szego, E. M., Bahr, M., Weishaupt, J. H. and
Lingor, P. (2012). "Inhibition of rho kinase enhances survival of dopaminergic neurons and
attenuates axonal loss in a mouse model of Parkinson's disease." Brain 135(Pt 11): 33553370.
Tonges, L., Gunther, R., Suhr, M., Jansen, J., Balck, A., Saal, K. A., Barski, E., Nientied, T., Gotz, A. A.,
Koch, J. C., Mueller, B. K., Weishaupt, J. H., Sereda, M. W., Hanisch, U. K., Bahr, M. and
Lingor, P. (2014). "Rho kinase inhibition modulates microglia activation and improves survival
in a model of amyotrophic lateral sclerosis." Glia 62(2): 217-232.
Tonges, L., Koch, J. C., Bahr, M. and Lingor, P. (2011). "ROCKing Regeneration: Rho Kinase Inhibition
as Molecular Target for Neurorestoration." Front Mol Neurosci 4: 39.
Toshima, Y., Satoh, S., Ikegaki, I. and Asano, T. (2000). "A new model of cerebral microthrombosis in
rats and the neuroprotective effect of a Rho-kinase inhibitor." Stroke 31(9): 2245-2250.
Tsukita, S. and Yonemura, S. (1997). "ERM proteins: head-to-tail regulation of actin-plasma
membrane interaction." Trends Biochem Sci 22(2): 53-58.
Tura, A., Schuettauf, F., Monnier, P. P., Bartz-Schmidt, K. U. and Henke-Fahle, S. (2009). "Efficacy of
Rho-kinase inhibition in promoting cell survival and reducing reactive gliosis in the rodent
retina." Invest Ophthalmol Vis Sci 50(1): 452-461.
Uehata, M., Ishizaki, T., Satoh, H., Ono, T., Kawahara, T., Morishita, T., Tamakawa, H., Yamagami, K.,
Inui, J., Maekawa, M. and Narumiya, S. (1997). "Calcium sensitization of smooth muscle
mediated by a Rho-associated protein kinase in hypertension." Nature 389(6654): 990-994.
Van de Velde, S., Van Bergen, T., Sijnave, D., Hollanders, K., Castermans, K., Defert, O., Leysen, D.,
Vandewalle, E., Moons, L. and Stalmans, I. (2014b). "AMA0076, a novel, locally acting Rho
kinase inhibitor, potently lowers intraocular pressure in New Zealand white rabbits with
minimal hyperemia." Invest Ophthalmol Vis Sci 55(2): 1006-1016.
Van Veldhuisen, P. C., Ederer, F., Gaasterland, D. E., Sullivan, K., Beck, A., Prum, B. E., Cyrlin, M. N.
and Weiss, H. (2000). "The Advanced Glaucoma Intervention Study (AGIS): 7. The relationship
between control of intraocular pressure and visual field deterioration. ." Am J Ophthalmol
130(4): 429-440.
Villar-Cheda, B., Dominguez-Meijide, A., Joglar, B., Rodriguez-Perez, A. I., Guerra, M. J. and
Labandeira-Garcia, J. L. (2012). "Involvement of microglial RhoA/Rho-kinase pathway
activation in the dopaminergic neuron death. Role of angiotensin via angiotensin type 1
receptors." Neurobiol Dis 47(2): 268-279.
Waki, M., Yoshida, Y., Oka, T. and Azuma, M. (2001). "Reduction of intraocular pressure by topical
administration of an inhibitor of the Rho-associated protein kinase." Curr Eye Res 22(6): 470474.
Walker, K. L., Yoo, H. K., Undamatla, J. and Szaro, B. G. (2001). "Loss of neurofilaments alters axonal
growth dynamics." J Neurosci 21(24): 9655-9666.
Wang, J. J., Mitchell, P. and Smith, W. (1997). "Is there an association between migraine headache
and open-angle glaucoma? Findings from the Blue Mountains Eye Study." Ophthalmology
104(10): 1714-1719.
Weibel, D., Kreutzberg, G. W. and Schwab, M. E. (1995). "Brain-derived neurotrophic factor (BDNF)
prevents lesion-induced axonal die-back in young rat optic nerve." Brain Res 679(2): 249-254.
Weinreb, R. N. (2007). "Glaucoma neuroprotection: What is it? Why is it needed?" Can J Ophthalmol
42(3): 396-398.
48
Weinreb, R. N. and Khaw, P. T. (2004). "Primary open-angle glaucoma." Lancet 363(9422): 17111720.
Weise, J., Isenmann, S., Klocker, N., Kugler, S., Hirsch, S., Gravel, C. and Bahr, M. (2000). "Adenovirusmediated expression of ciliary neurotrophic factor (CNTF) rescues axotomized rat retinal
ganglion cells but does not support axonal regeneration in vivo." Neurobiol Dis 7(3): 212-223.
Wiederholt, M., Thieme, H. and Stumpff, F. (2000). "The regulation of trabecular meshwork and
ciliary muscle contractility." Prog Retin Eye Res 19(3): 271-295.
Wierzbowska, J., Robaszkiewicz, J., Figurska, M. and Stankiewicz, A. (2010). "Future possibilities in
glaucoma therapy." Med Sci Monit 16(11): RA252-259.
Wolfs, R. C., Klaver, C. C., Ramrattan, R. S., van Duijn, C. M., Hofman, A. and de Jong, P. T. (1998).
"Genetic risk of primary open-angle glaucoma. Population-based familial aggregation study."
Arch Ophthalmol 116(12): 1640-1645.
Yamamoto, K., Maruyama, K., Himori, N., Omodaka, K., Yokoyama, Y., Shiga, Y., Morin, R. and
Nakazawa, T. (2014). "The novel Rho kinase (ROCK) inhibitor K-115: a new candidate drug for
neuroprotective treatment in glaucoma." Invest Ophthalmol Vis Sci.
Yamashita, T. and Tohyama, M. (2003). "The p75 receptor acts as a displacement factor that releases
Rho from Rho-GDI." Nat Neurosci 6(5): 461-467.
Yan, D. B., Coloma, F. M., Metheetrairut, A., Trope, G. E., Heathcote, J. G. and Ethier, C. R. (1994).
"Deformation of the lamina cribrosa by elevated intraocular pressure." Br J Ophthalmol
78(8): 643-648.
Yanagi, M., Kawasaki, R., Wang, J. J., Wong, T. Y., Crowston, J. and Kiuchi, Y. (2011). "Vascular risk
factors in glaucoma: a review." Clin Experiment Ophthalmol 39(3): 252-258.
Zhang, C. W., Lu, Q., You, S. W., Zhi, Y., Yip, H. K., Wu, W., So, K. F. and Cui, Q. (2005). "CNTF and
BDNF have similar effects on retinal ganglion cell survival but differential effects on nitric
oxide synthase expression soon after optic nerve injury." Invest Ophthalmol Vis Sci 46(4):
1497-1503.
Zhang, Z., Ottens, A. K., Larner, S. F., Kobeissy, F. H., Williams, M. L., Hayes, R. L. and Wang, K. K.
(2006). "Direct Rho-associated kinase inhibition [correction of inhibiton] induces cofilin
dephosphorylation and neurite outgrowth in PC-12 cells." Cell Mol Biol Lett 11(1): 12-29.
Zhao, J., Zhou, D., Guo, J., Ren, Z., Zhou, L., Wang, S., Xu, B. and Wang, R. (2006). "Effect of Fasudil
Hydrochloride, a Protein Kinase Inhibitor, on Cerebral Vasospasm and Delayed Cerebral
Ischemic Symptoms After Aneurysmal Subarachnoid Hemorrhage." Neurol Med Chir 46(9):
421-428.
Zhou, Y., Huang, X., Hecker, L., Kurundkar, D., Kurundkar, A., Liu, H., Jin, T. H., Desai, L., Bernard, K.
and Thannickal, V. J. (2013). "Inhibition of mechanosensitive signaling in myofibroblasts
ameliorates experimental pulmonary fibrosis." J Clin Invest 123(3): 1096-1108.
49
Download