GSK3 in Retinal Degenerative Diseases: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Glycogen synthase kinase 3 (GSK3) is a key regulator of many cellular signaling processes and performs a wide range of biological functions in the nervous system. Due to its central role in numerous cellular processes involved in cell degeneration, a rising number of studies have highlighted the interest in developing therapeutics targeting GSK3 to treat neurodegenerative diseases. GSK3 is a key player in retinal neuronal death in various retinal diseases. 

  • GSK3
  • retina
  • degeneration

1. GSK3 and Retinal Inflammation

Retinal inflammation is well known to contribute to the pathogenesis of retinal diseases, such as AMD, DR, or RP [138,139,140,141]. Inflammation is one of the important functions that are regulated by GSK3 since it is well established that GSK3 acts as a modulator of inflammatory components [38,142,143]. For instance, GSK3 negatively regulates anti-inflammatory cytokine production such as IL-2, IL-10, IL-22, or IL-33 [144]. Conversely, this kinase acts as a positive regulator of pro-inflammatory cytokines and chemokines, such as TNF-α, interleukin (IL-)1β, IL-6, IL-17, IL-18, IL-23, IL-12, IFN-γ, IL-8, C-C motif chemokine ligand (CCL) 2, 3, 4, and 12, C-X-C motif chemokine ligand (CXCL) 1, 2, 5, and 10 [144]. Together, this raised the possibility that inhibitors of GSK3 may prove to be beneficial for inflammatory conditions. One signaling molecule of interest targeted by GSK3 is NFκB, a pivotal mediator of inflammatory responses that has long been proposed as a potential target for the therapy of inflammatory diseases [145,146]. It was shown that NF-κB is activated in rd mice and light-induced retinal degeneration [147,148] Interestingly, GSK3β was shown to facilitate NF-κB transactivation by TNF-α since GSK3β deficient mouse embryonic fibroblasts exhibit defective NF-κB activation in response to TNFα [66]. Mechanistically, direct phosphorylation of NF-κB subunits p65 by GSK3 was reported in hepatocytes [149]. Along the same line, GSK3 inhibition in microglial cells decreases LPS-induced inflammation through the decrease of the activation of p65 [150]. GSK3 also activates non-canonical NF-κB signaling through the phosphorylation of p100, an inhibitor of NF-κB, targeting it to the proteasome [151]. Moreover, the increased β-catenin levels following GSK3 inactivation may further enhance NF-κB inhibition, since β-catenin was shown to inhibit NF-κB activity through physical interaction [152]. It is however noteworthy that GSK3 could differentially regulate NF-κB activity depending on the physiological state of the cell. Indeed, although it is required for the activation of NF-κB in response to cytokine stimulation [153], it may inhibit NF-κB in resting cultured cells [153,154,155]. Moreover, NF-κB can exhibit anti-apoptotic effects. Indeed, mice with inactivated GSK3β die from hepatocyte apoptosis during development due to a defect in NF-κB activation [66]. Finally, although GSK3 inhibition in different models predominantly contributes to the amelioration of inflammation, it may alternatively lead to the opposite effect as it could also prohibit the termination of inflammation [144]. As a whole, it is clear that more knowledge is needed to evaluate the net outcome of GSK3 inhibitors on NF-κB activity and on neuroinflammation in general in different models of retinal degeneration.
Another interesting factor linked to inflammation and GSK3 is the P2X7 receptor (P2X7-R) which is known to promote chronic neuroinflammation and neurodegenerative brain diseases [156]. In AD mouse models, P2X7-R inhibition was shown to have a protective effect through GSK3 inhibition [157,158]. P2X7-R is expressed in the retina and the RPE, and during retinal degeneration its expression increases [159]. P2X7-R induces the expression of inflammatory factors in the retina [160,161]. Interestingly, the pharmacological inhibition of P2X7-R prevents the increased inflammation and neovascularization induced by oxidative stress in the mouse eye in vivo [162]. It would be interesting to know whether some of these effects on inflammation and neuroprotection involve GSK3 inhibition.

2. GSK3 and Retinal Vascularization

Retinal neovascularization is observed in some retinal degenerative diseases, such as DR or wet AMD, where there is a disruption of the blood-retinal barrier (BRB). This phenotype is correlated with the increased expression of Vascular Endothelial Growth Factor (VEGF), a target gene of Wnt signaling [163,164]. A commonly used treatment strategy for wet-AMD and DR relies on reducing angiogenesis via anti-VEGF agents [165,166,167,168,169,170,171].
Wnt signaling activation promotes retinal vascularization [172,173,174], BRB development, and maintenance [175]. Accordingly, the reduction of Wnt signaling by the loss of LRP5, a canonical Wnt co-receptor, suppresses pathologic neovessel formation in a mouse model of oxygen-induced retinopathy [176]. Even if the loss of LRP5 negatively regulates retinal neovascularization in development and adulthood, there are still some vessels formed, with great disorganization and blood leakage [177]. Acting on downstream effectors of Wnt signaling could be another strategy to prevent neovascularization. In this pathological context, promoting GSK3 activity might thus be an interesting alternative approach to inhibit Wnt signaling and subsequently diminish VEGF expression.
In contrast to the occurrence of neovascularization in some retinal dystrophies, other eye diseases are characterized by the poor formation of intraocular vasculature, such as the familial exudative vitreoretinopathy (FEVR) [178]. Consistent with the importance of Wnt signaling in retinal vascular development, Wnt inhibition in Lrp5−/− mice produces eye vascular pathologies that model FEVR in humans [179]. Importantly, inhibition of GSK3 by LiCl treatment in this mouse model was shown to rescue defective retinal vasculature through restoring Wnt signaling, providing a potential treatment approach for FEVR [179].

3. GSK3 and Retinal Regeneration

Cell-based therapy is an appealing approach in late-stage retinal degeneration when most cells are already dead. One approach relies on the stimulation of endogenous repair processes. Some species have a high regenerative capacity, such as zebrafish or Xenopus, in which MGCs exhibit stemness properties. After retinal damage, dormant Müller cells can exit quiescence, proliferate and differentiate into different retinal cell types [180,181,182]. In contrast with fish and amphibians, these stemness and neurogenic capacities are highly limited in the mammalian retina [183,184]. The goal is to identify cellular pathways able in mammals to trigger Müller glia reprogramming and differentiation of Müller-derived progenitors under pathological conditions.
Several results gathered in different animal models converge on the idea that regeneration of retinal neurons can be promoted by applying GSK3 inhibitors to the retina. Zebrafish regenerative capacity observed after retinal damage is mediated by Ascl1 [185], which induces the Wnt signaling pathway [186]. Remarkably, in zebrafish undamaged retinas, Wnt activation, via GSK3 inhibition with LiCl, is sufficient to stimulate MGCs proliferation and induce retinal regeneration [186]. In the chick retina, which has a low regenerative potential, GSK3 drug-inhibition associated with FGF2 treatment promotes MGCs proliferation and dedifferentiation [187]. In the rat retina, although some MGCs are stimulated to proliferate and produce retinal cells in retinal explants, this remains very limited [188]. Activation of Wnt signaling, either by Wnt3a treatments or using SB216763 or AR-A014418 as inhibitors of GSK3β, was shown to promote the proliferation of Müller glia-derived retinal progenitors and neural regeneration in the wild-type retina [189] (Figure 1e). However, in rd mice, similar approaches induce MGC proliferation only at P12 but not at a later stage suggesting that the retinal microenvironment variation under pathological conditions might contribute to the lack of retinal repair observed in mammals [189]. Along the same line, the proliferative response of MGCs in mouse retinal explants following the addition of the GSK3 inhibitor Chir99021 varies between mouse strains, highlighting the importance of the genetic background [190]. Nevertheless, altogether these data in different models and species suggest that targeting GSK3 for retinal cell regeneration deserves further attention.
Given the critical function of GSK3 in regulating axon growth, modulation of GSK3 activity may also represent an interesting strategy to trigger axon regeneration following injuries [7,191,192,193,194]. Interestingly enough, antagonist effects were observed on GSK3 inhibition and axon growth. GSK3 activation promotes peripheral nerve axon growth, whereas GSK3 inhibition promotes CNS axon growth [195]. Lack of phosphatase and tensin homolog expression (Pten−/−) is neuroprotective and enhances RGC axon regeneration. Indeed, in this model, RGC axonal regeneration in mature neurons is promoted by activation of mTOR [196]. Such a mechanism relies essentially on GSK3 inactivation supporting that GSK3 inhibitors could serve as a regenerative stimulus.
Axon regeneration is an appealing approach for preserving the optic nerve in glaucoma [197]. Activation of Wnt signaling through intravitreal administration of Wnt3a after an optic nerve crush (ONC), a classical axon injury paradigm, contributes to axonal regeneration [198]. Consistent with this, GSK3β inhibition also enhances optic nerve regeneration after ONC [195]. This effect is mediated by the Collapsin response mediator protein 2 (CRMP2), a microtubule-binding protein involved in neuronal polarization, migration, and differentiation. Therefore, GSK3/CRMP2 axis is a pathway of interest to treat glaucoma by promoting axon regeneration [196]. Another interesting factor involved in regeneration is mTOR. mTOR positively regulates cell growth, proliferation, and survival, and this is mediated by activation of Wnt pathway, thus GSK3 inhibition [199]. Similarly, mTOR promotes axon regeneration in the CNS through GSK3 inhibition [200]. In the retina, after an ONC and inflammatory stimulation, mTOR signaling enhances optic nerve regeneration as well as RGCs neuroprotection [201]. Thus, inhibiting GSK3 is an appealing strategy for optic nerve regeneration as it should enhance the activity of key downstream effectors, in particular CRMP2 and mTOR.

4. Precautions and Advantages of Inhibiting GSK3 or GSK3 Targets as a Therapeutic Strategy for Eye Diseases

Retinal diseases are numerous but biological processes deregulated in each of them are often shared [11]. In this context, factors at the crossroad of multiple pathways offer great potential as therapeutic targets. Among them, GSK3 has been shown deregulated in multiple CNS disorders including retinal diseases and due to its central role in regulating multiple signaling pathways, both kinases and their downstream targets represent targets of choice (Table 1), especially in a nonstop growing aging population. However, several points must be considered with caution when considering GSK3 as a therapeutic target.
At first, it is important to consider the type of disease, its dynamics and its evolution. Indeed, using GSK3 inhibitors to modulate GSK3 activity should be finely regulated, depending on the disease and the disease’s stage. For the case of DR, GSK3 is reported as activated and its inhibition might be beneficial at the early stage of the disease to preserve RGCs [91] but then keeping GSK3 inactivated might promote VEGF expression and angiogenesis through downstream effectors of the Wnt pathway [172,173,174]. Along the same line, in AMD, GSK3 inhibition could be beneficial in the dry form when neovessels are absent, but deleterious in the wet form by promoting retinal neovascularization. One could speculate that the combination of GSK3 inhibitors with other drugs, such as anti-VEGF, could at the same time inhibit angiogenesis and enhance cell survival. Only after such a thorough analysis of GSK3 regulation and its implication in the disease progression can the use of a GSK3 inhibitor be considered. This also raises the question of whether GSK3 inhibition should be transient or chronic. Along the same line, regeneration could be initiated and enhanced by transient inhibition of GSK3 to induce MGC proliferation but might not be required thereafter, to avoid over-proliferation or allow further differentiation. Overall, the time window and the duration of the treatment must be fine-tuned and defined for each disease, for a specific stage of the disease, and/or for a particular cell type.
One more thing to consider is that GSK3 is ubiquitously expressed and presents different functions across the different cell types/tissues. The drug effect might not have the same efficacy whether it is administered in a systemic way or in a tissue-selective manner by local treatment. In contrast with the brain, the eye is easy to access, therefore intravitreal injection is often preferred. As such, it solves one of the issues of the pharmacological treatment targeting GSK3 activity and the ability of these drugs to cross the retinal blood barrier. This type of injection is commonly used in ophthalmology for treating wet forms of AMD by repeated injection of anti-VEGF to reduce the neovascularization occurring in this form and therefore delay photoreceptor degeneration. Therefore, an effort should be put into the research of carriers providing local delivery of the product using intravitreal injection. To achieve such local and long-term delivery of the drugs, liposomes or polymeric nanoparticles could be a solution as a drug carrier [214].
Yet, another point to be resolved is to know if both isozymes should be targeted regarding their high degree of functional redundancy to maximize the effects. Of note, most used inhibitors are targeting GSK3β, but it might also affect GSK3α. Regarding the literature, inhibition of both isozymes might be favorable [41]. Another unresolved issue nowadays is to precisely distinguish the role of each GSK3 isozyme in each pathology, depending on the stage of the disease.
Another strategy could be to target downstream pathways and substrates of GSK3, known to be involved in the pathology and allowing a neuroprotection through specific inhibition or activation. Indeed, due to the broad spectrum of GSK3 cellular targets (over 100 known substrates), its inhibition in the brain by the means of small molecules logically leads to many side effects and as a consequence few GSK3 inhibitors have reached phase 2 clinical trials. Therefore, a better strategy for more effective therapies could instead target specific GSK3 downstream targets. For instance, some promising treatments targeting GSK3-regulated pathways are combining anti-oxidant effects and anti-inflammatory effects, as is the case of flavonoids coming from fruits and vegetables [215,216] or cannabidiol [217]. To illustrate the interest in targeting GSK3 targets, one can cite NRF2 for its antioxidant and anti-inflammatory functions. Several studies demonstrated the therapeutic interest of overexpressing this factor using AAV vectors. NRF2-based gene therapy showed neuroprotective effects against oxidative stress [218]. Interestingly, a ganglion-cell-specific promoter Mcp-1 was used as it is expressed only in stressed RGCs, avoiding non-cell-specific effects. NRF2 gene therapy showed also neuroprotective effects in a mouse model of AMD using light damage [219]. Another GSK3 target relevant for RGCs preservation is CRMP2. A gene therapy approach in rats based on the expression of a constitutively active form of CRMP2 resulted in RGCs neuroprotection after an optic nerve injury [220]. An additional promising target discussed above is mTOR. Upregulation of mTOR by inhibition of upstream effectors, such as PTEN and TSC2, has a positive role in optic nerve regrowth [221]. Interestingly, in the cases of DR and wet AMD, inhibition of mTOR using drug inhibitors or AAV seems to be a good option to reduce vascularization by preserving endothelial structure [222,223,224]. Altogether, these preclinical results clearly demonstrated the therapeutic interest in targeting GSK3 substrates.
Overall, targeting GSK3 activity is challenging as a therapeutic approach but shows high potential. The challenge would be to target specific cell types for each disease and to determine whether the activation or inactivation should be transient or chronic. In this context, a better understanding of GSK3 function in different retinal diseases and identification of their targets will certainly help in developing new therapeutic approaches.

This entry is adapted from the peer-reviewed paper 10.3390/cells11182898

This entry is offline, you can click here to edit this entry!
Video Production Service