Skip to main content
Erschienen in: Molecular Neurodegeneration 1/2018

Open Access 01.12.2018 | Research article

Modulation of GSK-3 provides cellular and functional neuroprotection in the rd10 mouse model of retinitis pigmentosa

verfasst von: Alonso Sánchez-Cruz, Beatriz Villarejo-Zori, Miguel Marchena, Josefa Zaldivar-Díez, Valle Palomo, Carmen Gil, Ignacio Lizasoain, Pedro de la Villa, Ana Martínez, Enrique J. de la Rosa, Catalina Hernández-Sánchez

Erschienen in: Molecular Neurodegeneration | Ausgabe 1/2018

Abstract

Background

Retinitis pigmentosa (RP) is a group of hereditary retinal neurodegenerative conditions characterized by primary dysfunction and death of photoreceptor cells, resulting in visual loss and, eventually, blindness. To date, no effective therapies have been transferred to clinic. Given the diverse genetic etiology of RP, targeting common cellular and molecular retinal alterations has emerged as a potential therapeutic strategy.

Methods

Using the Pde6b rd10/rd10 mouse model of RP, we investigated the effects of daily intraperitoneal administration of VP3.15, a small-molecule heterocyclic GSK-3 inhibitor. Gene expression was analyzed by quantitative PCR and protein expression and phosphorylation by Western blot. Photoreceptor preservation was evaluated by histological analysis and visual function was assessed by electroretinography.

Results

In rd10 retinas, increased expression of pro-inflammatory markers and reactive gliosis coincided with the early stages of retinal degeneration. Compared with wild-type controls, GSK-3β expression (mRNA and protein) remained unchanged during the retinal degeneration period. However, levels of GSK-3βSer9 and its regulator AktSer473 were increased in rd10 versus wild-type retinas. In vivo administration of VP3.15 reduced photoreceptor cell loss and preserved visual function. This neuroprotective effect was accompanied by a decrease in the expression of neuroinflammatory markers.

Conclusions

These results provide proof of concept of the therapeutic potential of VP3.15 for the treatment of retinal neurodegenerative conditions in general, and RP in particular.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13024-018-0251-y) contains supplementary material, which is available to authorized users.
Abkürzungen
CD11B
Cluster of differentiation molecule 11B
CD68
Cluster of differentiation 68
CNS
Central nervous system
ERG
Electroretinographic recording
GCL
Ganglion cell layer
GFAP
Glial fibrillary acidic protein
GSK
Glycogen synthase kinase
IBA1
Ionized calcium-binding adapter molecule 1
IC50
Half maximal inhibitory concentration
IL1-β
Interleukin-1β
INL
Inner nuclear layer
NF-κB
Nuclear factor kappa-light-chain-enhancer of activated B cells
ON
Optic nerve
ONL
Outer nuclear layer
OP
Oscillatory potential
OS
Outer segment
PB
Phosphate buffer
PDE
Phosphodiesterase
PKA
Protein kinase A
PKCα
Protein kinase C alpha
qPCR
Quantitative polymerase chain reaction
RBPMS
RNA binding protein with multiple splicing
RP
Retinitis pigmentosa
RT
Reverse transcription
TBP
TATA-binding protein
TNFα
Tumor necrosis factorα
WT
Wild-type
α2M
α-2-macroglobulin

Background

Inherited retinal dystrophies, which include retinitis pigmentosa (RP), are a group of genetic diseases caused by mutations in over 300 genes and loci (http://​www.​sph.​uth.​tmc.​edu/​Retnet/​disease.​htm). RP is a retinal neurodegenerative condition characterized by primary dysfunction and death of photoreceptor cells, resulting in vision loss and, ultimately, blindness [1]. Although several experimental therapies have advanced to clinical trials (https://​clinicaltrials.​gov/​ct2/​results?​term=​retinitis+pigmen​tosa&​Search=​Search), neither approved treatments nor preventive therapies for RP are currently available. Its status as a rare disease and its diverse genetic etiology emphasize the importance of identifying shared pathological mechanisms independent of the causative mutation. Targeting common cellular and molecular retinal responses to mutations could benefit a significant number of RP patients, as well as those with other retinal dystrophies without an exclusively genetic etiology (e.g., glaucoma, age-related macular degeneration, diabetic retinopathy). Recent studies suggest that retinal neurodegeneration is associated with a broad inflammatory response in the retina. This response appears to be mutation-independent and involves microglial activation, reactive macrogliosis, and the production of pro-inflammatory cytokines [25].
GSK-3 is a serine/threonine kinase that exists as 2 highly homologous isoforms, GSK-3α and GSK-3β, each encoded by a distinct gene. GSK-3β is predominantly expressed in the central nervous system (CNS) [6]. Although initially identified as a glycogen synthesis enzyme (the role for which it was named), GSK-3 is currently considered a multitask enzyme involved in the regulation of diverse cellular functions owing to its broad substrate spectrum [7, 8]. In particular, GSK-3 plays a pivotal role in regulating the balance between pro-inflammatory and anti-inflammatory cellular responses. Consequently, GSK-3 is considered a potential therapeutic target for diseases with an inflammatory component. The therapeutic potential of GSK-3 modulators is currently being studied in a variety of neuroinflammatory diseases. These include psychiatric and neurodegenerative disorders, as well as retinal dystrophies; as part of the CNS, the retina shares many physiological and pathological traits with the brain [9].
Employing a chemical genetic approach, we recently investigated the neuroprotective effects of 3 chemically diverse GSK-3 modulators in photoreceptor cells [10]. In the present study we selected one of these compounds, VP3.15, based on its favorable pharmacokinetic and IC50 properties. This molecule is a 5-imino-thiadiazole that has been described not only as substrate-competitive GSK-3 inhibitor [11], but also as allosteric inhibitor of PDE7 [12].
We sought to validate in vivo the potential of GSK-3 as a therapeutic target for the treatment of RP, and to demonstrate the therapeutic potential of VP3.15 in this neurodegenerative condition. We characterized the expression of GSK-3β and its inactive serine-9-phosphorylated form in the dystrophic retina of the rd10 mouse, a model of RP. Moreover, we demonstrated that chronic systemic in vivo VP3.15 treatment preserved visual function by delaying neurodegeneration. Our results indicate that VP3.15 is an innovative drug candidate for the treatment of RP.

Methods

Animals and drug delivery

The rd10 mouse model of retinal neurodegeneration carries a homozygous phosphodiesterase 6b mutation (Pde6b rd10/rd10 ) on a C57BL/6 J background. Mice were kindly provided by Bo Chang from The Jackson Laboratory (Bar Harbor, ME, USA). Wild type (WT) control mice of the same background were also obtained from The Jackson Laboratory. All animals were housed and handled in accordance with the ARVO statement for the Use of Animals in Ophthalmic and Vision Research, European Union guidelines, and those of the local ethics committees of the CSIC and the Comunidad de Madrid. Mice were bred at the CIB core facilities on a 12/12-h light/dark cycle.
VP3.15 was synthesized in our laboratory as previously described [11]. This small molecule is a member of the iminothiadiazole family, the first group of substrate-competitive GSK-3 inhibitors described ([11]; see Additional file 1: Figure S1 for the structure data). The dose administrated was selected based on pharmacokinetic and IC50 data ([1113] and data not shown). Mice received daily intraperitoneal injections of vehicle (10% DMSO, 0.9% NaCl) or VP3.15 (10 mg/kg) for the indicated period of time.
IP administration was selected based on the ability of VP3.15 to cross the brain blood barrier [13] and the necessity of performing repetitive administration without damaging the eye.

Electroretinography

Mice underwent electroretinographic recordings (ERG) at different time points, using a previously described ERG protocol [14]. ERGs were performed by an observer blind to treatment. ERG signals were amplified and band filtered between 0.3 Hz and 1000 Hz (CP511 Preamplifier, Grass Instruments, Quincy, MA, USA) and digitized at 10 kHz using a PowerLab acquisition data card (AD Instruments Ltd., Oxfordshire, UK). Graphical representations of the signals recorded and the control light stimuli were generated using Scope v6.4 PowerLab software. ERG wave amplitudes were measured off-line and the results averaged.

Histology and immunostaining

Mice were euthanized at the indicated ages and their eyes enucleated. The right eye was processed for histological analyses and the left eye for retinal RNA extraction. For histological analyses, eyes were fixed for 1 h in 4% (w/v) paraformaldehyde in 0.1 M phosphate buffer (PB), pH 7.4, and then cryoprotected by incubation in increasing concentrations of sucrose (final concentration 50% (w/v) in PB). The eyes were then embedded in Tissue-Tek OCT (Sakura Finetec, Torrance, CA, USA) and frozen on isopropanol/dry ice. Cryostat equatorial sections (12 μm) were mounted on Superfrost® Plus slides (Thermo Scientific, Massachusetts, USA), dried at room temperature, and stored at − 20 °C. Before performing further analyses, sections were fixed in acetone for 10 min at − 20 °C and dried at 65 °C for 10 min. After rinsing in PBS and permeation with 1% (w/v) Triton X-100 in PBS, sections were blocked in BGT (2.5 g/L BSA, 100 mM glycine, 0.25% (w/v) Triton X-100 in PBS) for 1 h and then incubated overnight at 4 °C with primary antibodies (Table 1) diluted in BGT. Sections incubated in the absence of primary antibody were used as specificity controls. After rinsing in PBS and incubation with the appropriate secondary antibodies (Table 1), sections were stained with DAPI (4′,6-diamidino-2-phenylindole; Sigma-Aldrich Corp., St. Louis, MO, USA) and coverslipped with Fluoromont-G.
Table 1
Antibodies
Antibody against
Host species
Dilution
Manufacturer
Catalog number
Rhodopsin
Mouse
IH,1:500
Abcam, Cambridge, UK
AB3267
L/M-opsin
Rabbit
IH,1:200
Abcam
AB5405
Mouse-Igs, Rabbit-Igs Alexa 488-546-647 labeled
Goat
IH, 1:200–500
ThermoFisher Scientific, Waltham, MA
A-11001
A-11008
A-11004
A-11011
A-21235
GSK-3Ββ
Mouse
IH,1:100
Cell Signaling Technology, Danvers, Massachusetts, USA
9832S
pGSK-3ΒβS9
Mouse
IH,1:400
Cell Signaling Technology
93,235
Recoverin
Rabbit
WB,1:5000
Millipore, Billerica, Massachusetts, USA
AB5585
GSK-3Ββ
Mouse
WB,1:1000
Cell Signaling Technology
9832S
pGSK-3ΒβS9
Rabbit
WB,1:1000
Cell Signaling Technology
93,235
Akt
Rabbit
WB,1:1000
Cell Signaling Technology
9272
pAktS473
Rabbit
WB,1:1000
Cell Signaling Technology
9271 L
pAktS473
Rabbit
IH,1:100
Cell Signaling Technology
4060
GFAP
Rabbit
IH, 1:500
Dako, Glostrup (Denmark)
Z0334
IH immunohistochemistry, WB Western blot
To assess preservation of the photoreceptor layer, we compared the thickness of the ONL (which primarily contains photoreceptors) with that of the corresponding INL (which contains bipolar, horizontal, and amacrine neurons and Müller glial cell bodies), and quantified the length of rod and cone outer segments (OS). Three sections per eye were analyzed: for each section, one image was captured for each of the 6 retinal zones (T1, T2, T3, T4, T5, and T6; Additional file 2: Figure S2). In each image, 3 measurements were recorded at random positions to obtain an average value per retinal zone per section. Measurements were performed using the “freehand line” and “measure” tools in Fiji software. The ONL thickness was normalized to that of the INL (not affected by the degeneration at this stage) to correct for possible inclinations of the sectioning plane.

Immunoblots

Protein extraction was carried out by sonicating individual retinas in RIPA lysis buffer (containing 2 mM Na3VO4, 10 mM NaF, and 4 mM Na4P2O7) and chilling the resulting solution on ice for 30 min. Protein (30 μg) from each sample was fractionated by electrophoresis on precast 10–12% (w/v) SDS-polyacrylamide gels (Criterion TGX, Bio-Rad, Munich, Germany), after which proteins were transferred to PVDF membranes using the Trans-Blot Turbo system (Bio-Rad). Blots were incubated overnight at 4 °C with primary antibodies (Table 1) diluted in TBS (Tris-buffered saline) containing 1% (w/v) Triton-X100, followed by incubation with the appropriate peroxidase-conjugated secondary antibody (Table 1). Proteins were visualized using the Pierce® ECL Western Blotting Substrate (ThermoFisher Scientific, Waltham, MA, USA), and quantified using ChemiDoc™ Touch Imaging System (Bio-Rad).

RNA isolation and quantitative PCR

Total RNA from individual retinas was extracted using TRIzol Reagent, and 2.5 μg of RNA was typically reverse transcribed using the Superscript III Kit and random primers (all from ThermoFisher Scientific). Quantitative PCR (qPCR) was performed with the ABI Prism 7900HT Sequence Detection System using TaqMan Universal PCR Master Mix, no-AmpErase UNG, and Taqman assays (listed below) for detection (all from Thermo Fisher). The relative change in gene expression was calculated using the 2ΔCt method, normalizing to expression levels of the Tbp (TATA-binding protein) gene. The primer-probe sets used are listed in Table 2.
Table 2
Taqman assays
Gene
Taqman assay
α2m
Mm00558642_m1
Gfap
Mm01253033_m1
Il1β
Mm00434228_m1
Tbp
Mm00446971_m1
Tnfα
Mm00443260_g1
Rho
Mm01184405_m1
Iba1
Mm00479862_g1
Cd68
Mm03047343_m1
Cd11b
Mm00434455_m1

Statistical analysis

Statistical analyses were performed with GraphPad Prism software 6.0 (GraphPad Software Inc., La Jolla, CA, USA). Protein levels were compared using an unpaired t-test. Gene expression levels were compared using either one-way or 2-way ANOVA tests as indicated in the Figure Legend. For ERG data, differences in wave amplitudes over time between vehicle- and VP3.15-treated mice were assessed using a 2-way ANOVA. Histological differences between vehicle- and VP3.15-treated mice were assessed by 2-way ANOVA. In all cases, p-values ≤0.05 were considered statistically significant.

Results

Inflammatory genes are upregulated in rd10 mice during the early stages of retinal neurodegeneration

Pathological neuroinflammation is a hallmark of neurodegeneration in many retinal dystrophies [25]. We analyzed temporal alterations in pro-inflammatory gene expression in the rd10 retina between P14 (before the appearance of morphological signs of retinal degeneration) and P21 (at which stage the degenerative process has become clearly established) to identify the optimal period for therapeutic intervention. Quantitative PCR analysis of WT and rd10 retinas revealed increased expression of the pro-inflammatory markers α2m, Il1β and Tnfα in the degenerating retinas, beginning in the early stages of degeneration (P18) and increasing dramatically (by 25 to 50 fold) by P21 (Fig. 1). Reactive gliosis in response to retinal damage, as measured by upregulated Gfap expression, correlated with the increased expression of inflammatory markers. Likewise, the expression of the microglia genes Cd68 and Cd11b also showed a 4–5 fold increase (Fig. 1).

GSK-3β expression in wild type and rd10 retinas

We next determined expression levels of GSK-3β, the most abundant GSK-3 isoform in the CNS [6], during the same degenerative period as above. We measured the expression of total GSK-3β and of its inactive Ser9-phosphorylated form (GSK-3βSer9) in rd10 mouse retinas. No significant differences in GSK-3β RNA levels were observed between rd10 and WT retinas (Fig. 2a). Furthermore, despite the morphological differences caused by the degenerative process, we observed similarly broad distribution patterns for GSK-3β protein in both P21 rd10 and WT retinas (Fig. 2b).
GSK-3β is an unusual kinase in that it is constitutively activated but is inhibited upon stimulation of its regulatory signaling pathways (reviewed in [7]). Activation of the PI3K/Akt pathway induces phosphorylation of GSK-3β at Ser9, and subsequent downregulation of GSK-3β activity [15]. During early retinal degeneration (P16 and P18), no differences in Ser473-phosphorylated Akt (pAktSer473) or Ser9-phosphorylated GSK-3β (pGSK-3βSer9) levels were observed between WT and rd10 retinas (data not shown). Conversely, at P19 and P21, at which point the degenerative process is well established, as evidenced by decreased expression of the photoreceptor protein recoverin, pAktSer473 and pGSK-3βSer9 levels were significantly higher in rd10 versus WT retinas (Fig. 3a and b). These observations in retinal extracts were in agreement with the immunostaining pattern observed at P21, which showed an increment in pAktSer473 and pGSK-3βSer9 in the rd10 retina (Fig. 3c). This inhibitory phosphorylation of GSK-3β, which coincided with activation of the pro-survival Akt signaling pathway, may represent an intrinsic neuroprotective response to the genetic damage caused in the retina. To test this hypothesis, we sought to potentiate this intrinsic response by pharmacologically inhibiting GSK-3 activity.

In vivo VP3.15 treatment decreases inflammatory and degenerative signs in the rd10 mouse retina

In light of our findings concerning proinflammatory gene expression and GSK-3β modulation, we selected the P15–P21 interval to assess the effects of short-term treatment with VP3.15, a GSK-3 inhibitor. VP3.15 inhibitory activity on GSK-3 was confirmed by determining its effect on the β-catenin levels in the microglia cell line N9 (Additional file 3: Figure S3). rd10 littermates received daily intraperitoneal injections of either vehicle or VP3.15 (10 mg/kg). One eye from each animal was analyzed for gene expression and the other for retinal morphology. VP3.15 treatment significantly decreased the expression of the proinflammatory genes Il1β and α2m. A similar trend was observed for Tnfα expression (Fig. 4a), and secretion by rd10 retinal explants (Additional file 7: Figure S7b), although these failed to reach statistical significance. The microglia genes Cd11b and Iba1 were also decreased upon VP3.15 treatment, while the activated microglia marker Cd68 showed a decline trend (Fig. 4b). In parallel, VP3.15 significantly reduced the expression of Gfap, an effect that correlates with a reduced GFAP staining depicted by the radial process of Müller glial cells and by the most inner retinal layer formed by Müller end feet and astrocytes (Fig. 4c and d). Conversely, a trend towards increased gene expression of rod photoreceptor-specific rhodopsin was observed in VP3.15-treated rd10 mice (Fig. 4e). This observation correlated with the maintenance of ONL thickness and OS length, both of which were better preserved in VP3.15-treated than in the vehicle-treated rd10 mice (Fig. 4f). Moreover, while rhodopsin expression was specifically localized in the photoreceptor outer segment (OS) in WT and VP3.15-treated rd10 retinas, vehicle-treated rd10 retinas displayed shortening of the OS and mislocalization of rhodopsin in the ONL (Fig. 4f).

In vivo treatment of rd10 mice with VP3.15 delays the loss of visual function

Based on the promising results obtained following short-term GSK-3 inhibition, we investigated whether a longer treatment period preserved visual function in the rd10 mouse model. rd10 littermates received daily injections of either vehicle or VP3.15 from P15 to P46 inclusive. Visual function was assessed weekly by ERG between P25 and P46. Compared with vehicle-treated counterparts, VP3.15-treated rd10 mice displayed better-defined ERG waves of greater amplitude (Fig. 5 and Additional file 4: Figure S4). The amplitude of ERG waves (a-mixed, b-mixed, b-photopic, and oscillatory potential [OP]) was significantly higher in VP3.15-treated than in vehicle-treated mice, indicating a prolongation of both rod- and cone-mediated vision function by VP3.15 (Fig. 5).
Histological evaluation of retinas at the end point of the study (P47, after the last ERG) revealed a greater relative ONL thickness in VP3.15- versus vehicle-treated retinas despite the advanced degeneration stage (Fig. 6a and b). The photoreceptor preservation effect of VP3.15 was more evident at mid-degeneration stages (P33; Additional file 5: Figure S5). Moreover, specific immunostaining showed better preservation of photoreceptor structure in VP3.15-treated retinas (Fig. 6a). Rhodopsin immunostaining in rod outer segments confirmed the partial preservation of rod outer segments in VP3.15-treated retinas. By contrast, rod outer segments were barely present in vehicle-treated retinas. Similarly, M-L opsin immunostaining revealed longer cone outer segments in VP3.15- versus vehicle-treated rd10 retinas (Fig. 6a and b).
Moreover, no toxic effect of VP3.15 administration was observed in other neuronal populations of the retina, particularly rod-bipolar and ganglion cells (Additional file 6: Figure S6).
Taken together, these results demonstrate a neuroprotective effect of VP3.15 on neuroinflammation, retinal structure, and visual function. Our findings thus support the potential of VP3.15 as a novel therapy for retinal dystrophies.

Discussion

This study describes GSK-3β expression and activation of the Akt-GSK-3β pathway during the early stages of retinal neurodegeneration in the rd10 mouse model of RP. Our findings support the involvement of GSK-3 in retinal decay and provide proof of concept of the neuroprotective effect of GSK-3 inhibition on photoreceptor cells [10]. In vivo administration of VP3.15, a small-molecule GSK-3 inhibitor, reduced photoreceptor cell loss and extended visual function. This neuroprotective effect was accompanied by a decrease in the expression of neuroinflammatory genes in the retina, indicating the potential of VP3.15 as a candidate RP therapy.
GSK-3 is a constitutively activated multitask enzyme, the activity of which is regulated by several inhibitory signaling pathways [7]. Inhibitory phosphorylation of GSK-3 at Ser9 is one of the main regulatory mechanisms. Our analysis of GSK-3β RNA and protein levels during the degenerative period in rd10 retinas revealed no differences with respect to WT counterparts. However, at both P19 and P21, by when photoreceptor degeneration is well established, inhibitory phosphorylation of GSK-3β at Ser9, and phosphorylation of its Akt regulator, were increased in rd10 retinas. This endogenous activation of the prosurvival Akt-GSK-3β pathway in the rd10 retina is in agreement with previous observations in a photoreceptor cell line subjected to a variety of insults, and in the dystrophic retina [16, 17]. We speculate that the activation of endogenous prosurvival pathways in the dystrophic retina constitutes an attempt to control the degenerative process, a response that, however, results insufficient to counteract the permanent genetic damage underlying the degeneration. In our experiments, administration of the GSK-3 inhibitor VP3.15 may have exogenously potentiated the prosurvival pathway, thus delaying photoreceptor cell death and preserving visual function. Indeed, several agents with neuroprotective effects in the retina exert common stimulatory effects on the Akt-GSK-3β pathway [1820]. GSK-3 is a downstream substrate of the insulin receptor [15, 21] and is thus inhibited upon insulin receptor stimulation. We have previously demonstrated that proinsulin, which binds with high affinity to the A isoform of the insulin receptor in the retina and activates Akt [22, 23], similarly prolongs rod survival and preserves visual function [14, 22, 24].
Neuroinflammation is a key component of neurodegenerative processes affecting the brain and retina, and thus constitutes a promising non-cell-autonomous target for treatments of CNS diseases in general, and of the retina in particular [25, 25]. GSK-3 plays a pivotal role in the regulation of peripheral and central pro-inflammatory cytokine production [26, 27], and its inhibition reduces both systemic and brain inflammation [26, 27]. The specific molecular events that occur in the rd10 retina upon GSK-3 inhibition remain to be established. This is a complicated task since GSK-3 is implicated in the regulation of multiple cellular processes including metabolism, cell structure, cell death, proliferation, and gene expression, with over 100 confirmed and 500 predicted substrates [7]. Exploratory studies employing rd10 retinal explants showed that VP3.15 attenuated NF-kB activation (Additional file 7: Figure S7). NF-kB is a key regulator of the inflammatory response by enabling the transcription of the genes encoding many pro-inflammatory cytokines. Indeed, our results suggest that levels of pro-inflammatory cytokines (Il1β and Tnfα) are reduced in VP3.15-treated rd10 retinas. This could explain, at least in part, the attenuation of photoreceptor degeneration and loss, and the preservation of vision. Moreover, the reduced inflammatory response observed in VP3.15-treated retinas was accompanied by decreases in the reactive gliosis marker GFAP as well as in α2M expression, for which a role in retinal neurodegeneration has been recently described [2830].
In addition to its inhibitory effect on GSK-3, VP3.15 also acts as an allosteric inhibitor of PDE7, an effect that contributes to its neuroprotective activity in other CNS pathologies [13]. However, PDE7 inhibition, via increasing cAMP levels and PKA activation, also converges on GSK-3 inhibition [31]. PKA is able to phosphorylate GSK-3β at Ser9 and, subsequently, to inactivate it [20, 31]. Besides, previous findings by our group support that the prosurvival effect of VP3.15 in photoreceptors may be primarily due to GSK-3 inhibition, since the specific GSK-3 inhibitor tideglusib exerted an even more potent neuroprotective effect in photoreceptors [10].
Most forms of RP involve primary death of rod photoreceptors, in which the mutated gene exerts its function, followed by secondary loss of cone photoreceptors. VP3.15 treatment prolonged photoreceptor survival, as evidenced by the preservation of ONL thickness and rod outer segment length. Furthermore, VP3.15 preserved the integrity of the cone outer segment, the first site of morphological alterations during cone degeneration. Human vision primarily relies on cones, which mediate daylight, color, and high-acuity vision. Accordingly, a mutation-independent treatment for RP that specifically prolongs cone survival would be highly beneficial. VP3.15 had a marked effect on cone cytoarchitecture and visual function, as indicated by an increase in the amplitude of the photopic b-wave compared with the untreated rd10 animals. It remains unclear whether VP3.15 exerts a direct effect on cone integrity, or whether this effect is secondary to the prosurvival effect on rods (present study and [10]). Regardless, VP3.15 resulted in a marked attenuation of the loss of both daylight and dim-light vision.

Conclusion

Our in vivo findings, together with the results of a recently published in vitro study [10], support the therapeutic potential of GSK-3 modulation for the treatment of retinal neurodegeneration, as described for other neurodegenerative diseases [32], and underscore the potential of GSK-3 inhibitors, in particular VP3.15, as pharmacological therapies for RP.

Acknowledgements

We thank Cayetana Murillo, Laura Ramírez and the staff of the CIB animal house and microscopy facilities for technical support. We thank Violeta Gómez-Vicente for advice on GSK-3 immunostaining. We thank Dr. García-Pardo and Dr. Labandeira-García for providing the pNF-κBser536 antibody and the N9 cell line, respectively. ASC and JZD are recipients of UCM and MECD (FPU13-00362) predoctoral fellowships, respectively.

Funding

This work was supported by grants from the Spanish MINECO (SAF2012-37979-C03-01 and SAF2016-76639-R to AM, SAF2013-41059-R and SAF2016-75681-R to EJdlR, and PI13-02098 to PdlV) and Instituto de Salud Carlos III and cofinanced by FEDER (PI17/01601 and RETICS RD16/0019/0009 to IL).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Ethics approval

Animal handling and experimental procedures followed the 3Rs principles, and were revised by the Animal Welfare Committee of the Centro de Investigaciones Biológicas (CIB) and the Bioethics Committee of the Consejo Superior de Investigaciones Científicas (CSIC) and authorized by the Comunidad de Madrid (Ref: PROEX 157/14), in accordance with the Spanish legislation and the European Union (EU) Directive 2010/63/EU for animal research.
All authors consent for the publication of this study.

Competing interests

CSIC has filed a patent for the VP3.15 (AM, VP and CG inventors).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Literatur
2.
Zurück zum Zitat Chinskey ND, Besirli CG, Zacks DN. Retinal cell death and current strategies in retinal neuroprotection. Curr Opin Ophthalmol. 2014;25:228–33.CrossRefPubMed Chinskey ND, Besirli CG, Zacks DN. Retinal cell death and current strategies in retinal neuroprotection. Curr Opin Ophthalmol. 2014;25:228–33.CrossRefPubMed
3.
Zurück zum Zitat Cuenca N, Fernandez-Sanchez L, Campello L, Maneu V, De la Villa P, Lax P, Pinilla I. Cellular responses following retinal injuries and therapeutic approaches for neurodegenerative diseases. Prog Retin Eye Res. 2014;43:17–75.CrossRefPubMed Cuenca N, Fernandez-Sanchez L, Campello L, Maneu V, De la Villa P, Lax P, Pinilla I. Cellular responses following retinal injuries and therapeutic approaches for neurodegenerative diseases. Prog Retin Eye Res. 2014;43:17–75.CrossRefPubMed
4.
Zurück zum Zitat Mustafi D, Maeda T, Kohno H, Nadeau JH, Palczewski K. Inflammatory priming predisposes mice to age-related retinal degeneration. J Clin Invest. 2012;122:2989–3001.CrossRefPubMedPubMedCentral Mustafi D, Maeda T, Kohno H, Nadeau JH, Palczewski K. Inflammatory priming predisposes mice to age-related retinal degeneration. J Clin Invest. 2012;122:2989–3001.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Yoshida N, Ikeda Y, Notomi S, Ishikawa K, Murakami Y, Hisatomi T, Enaida H, Ishibashi T. Clinical evidence of sustained chronic inflammatory reaction in retinitis pigmentosa. Ophthalmology. 2013;120:100–5.CrossRefPubMed Yoshida N, Ikeda Y, Notomi S, Ishikawa K, Murakami Y, Hisatomi T, Enaida H, Ishibashi T. Clinical evidence of sustained chronic inflammatory reaction in retinitis pigmentosa. Ophthalmology. 2013;120:100–5.CrossRefPubMed
6.
Zurück zum Zitat Pardo M, Abrial E, Jope RS, Beurel E. GSK3beta isoform-selective regulation of depression, memory and hippocampal cell proliferation. Genes Brain Behav. 2016;15:348–55.CrossRefPubMedPubMedCentral Pardo M, Abrial E, Jope RS, Beurel E. GSK3beta isoform-selective regulation of depression, memory and hippocampal cell proliferation. Genes Brain Behav. 2016;15:348–55.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Beurel E, Grieco SF, Jope RS. Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases. Pharmacol Ther. 2015;148:114–31.CrossRefPubMed Beurel E, Grieco SF, Jope RS. Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases. Pharmacol Ther. 2015;148:114–31.CrossRefPubMed
8.
Zurück zum Zitat Jope RS, Johnson GV. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 2004;29:95–102.CrossRefPubMed Jope RS, Johnson GV. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 2004;29:95–102.CrossRefPubMed
9.
Zurück zum Zitat Jindal V. Interconnection between brain and retinal neurodegenerations. Mol Neurobiol. 2015;51:885–92.CrossRefPubMed Jindal V. Interconnection between brain and retinal neurodegenerations. Mol Neurobiol. 2015;51:885–92.CrossRefPubMed
10.
Zurück zum Zitat Marchena M, Villarejo-Zori B, Zaldivar-Diez J, Palomo V, Gil C, Hernandez-Sanchez C, Martinez A, de la Rosa EJ. Small molecules targeting glycogen synthase kinase 3 as potential drug candidates for the treatment of retinitis pigmentosa. J Enzyme Inhib Med Chem. 2017;32:522–6.CrossRefPubMed Marchena M, Villarejo-Zori B, Zaldivar-Diez J, Palomo V, Gil C, Hernandez-Sanchez C, Martinez A, de la Rosa EJ. Small molecules targeting glycogen synthase kinase 3 as potential drug candidates for the treatment of retinitis pigmentosa. J Enzyme Inhib Med Chem. 2017;32:522–6.CrossRefPubMed
11.
Zurück zum Zitat Palomo V, Perez DI, Perez C, Morales-Garcia JA, Soteras I, Alonso-Gil S, Encinas A, Castro A, Campillo NE, Perez-Castillo A, et al. 5-imino-1,2,4-thiadiazoles: first small molecules as substrate competitive inhibitors of glycogen synthase kinase 3. J Med Chem. 2012;55:1645–61.CrossRefPubMed Palomo V, Perez DI, Perez C, Morales-Garcia JA, Soteras I, Alonso-Gil S, Encinas A, Castro A, Campillo NE, Perez-Castillo A, et al. 5-imino-1,2,4-thiadiazoles: first small molecules as substrate competitive inhibitors of glycogen synthase kinase 3. J Med Chem. 2012;55:1645–61.CrossRefPubMed
12.
Zurück zum Zitat Redondo M, Palomo V, Brea J, Perez DI, Martin-Alvarez R, Perez C, Paul-Fernandez N, Conde S, Cadavid MI, Loza MI, et al. Identification in silico and experimental validation of novel phosphodiesterase 7 inhibitors with efficacy in experimental autoimmune encephalomyelitis mice. ACS Chem Neurosci. 2012;3:793–803.CrossRefPubMedPubMedCentral Redondo M, Palomo V, Brea J, Perez DI, Martin-Alvarez R, Perez C, Paul-Fernandez N, Conde S, Cadavid MI, Loza MI, et al. Identification in silico and experimental validation of novel phosphodiesterase 7 inhibitors with efficacy in experimental autoimmune encephalomyelitis mice. ACS Chem Neurosci. 2012;3:793–803.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Medina-Rodriguez EM, Bribian A, Boyd A, Palomo V, Pastor J, Lagares A, Gil C, Martinez A, Williams A, de Castro F. Promoting in vivo remyelination with small molecules: a neuroreparative pharmacological treatment for multiple sclerosis. Sci Rep. 2017;7:43545.CrossRefPubMedPubMedCentral Medina-Rodriguez EM, Bribian A, Boyd A, Palomo V, Pastor J, Lagares A, Gil C, Martinez A, Williams A, de Castro F. Promoting in vivo remyelination with small molecules: a neuroreparative pharmacological treatment for multiple sclerosis. Sci Rep. 2017;7:43545.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Corrochano S, Barhoum R, Boya P, Arroba AI, Rodriguez-Muela N, Gomez-Vicente V, Bosch F, de Pablo F, de la Villa P, de la Rosa EJ. Attenuation of vision loss and delay in apoptosis of photoreceptors induced by proinsulin in a mouse model of retinitis pigmentosa. Invest Ophthalmol Vis Sci. 2008;49:4188–94.CrossRefPubMed Corrochano S, Barhoum R, Boya P, Arroba AI, Rodriguez-Muela N, Gomez-Vicente V, Bosch F, de Pablo F, de la Villa P, de la Rosa EJ. Attenuation of vision loss and delay in apoptosis of photoreceptors induced by proinsulin in a mouse model of retinitis pigmentosa. Invest Ophthalmol Vis Sci. 2008;49:4188–94.CrossRefPubMed
15.
Zurück zum Zitat Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995;378:785–9.CrossRefPubMed Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995;378:785–9.CrossRefPubMed
16.
Zurück zum Zitat Finnegan S, Mackey AM, Cotter TG. A stress survival response in retinal cells mediated through inhibition of the serine/threonine phosphatase PP2A. Eur J Neurosci. 2010;32:322–34.CrossRefPubMed Finnegan S, Mackey AM, Cotter TG. A stress survival response in retinal cells mediated through inhibition of the serine/threonine phosphatase PP2A. Eur J Neurosci. 2010;32:322–34.CrossRefPubMed
17.
Zurück zum Zitat Nakazawa T, Shimura M, Tomita H, Akiyama H, Yoshioka Y, Kudou H, Tamai M. Intrinsic activation of PI3K/Akt signaling pathway and its neuroprotective effect against retinal injury. Curr Eye Res. 2003;26:55–63.CrossRefPubMed Nakazawa T, Shimura M, Tomita H, Akiyama H, Yoshioka Y, Kudou H, Tamai M. Intrinsic activation of PI3K/Akt signaling pathway and its neuroprotective effect against retinal injury. Curr Eye Res. 2003;26:55–63.CrossRefPubMed
18.
Zurück zum Zitat Baek SM, Yu SY, Son Y, Hong HS. Substance P promotes the recovery of oxidative stress-damaged retinal pigmented epithelial cells by modulating Akt/GSK-3beta signaling. Mol Vis. 2016;22:1015–23.PubMedPubMedCentral Baek SM, Yu SY, Son Y, Hong HS. Substance P promotes the recovery of oxidative stress-damaged retinal pigmented epithelial cells by modulating Akt/GSK-3beta signaling. Mol Vis. 2016;22:1015–23.PubMedPubMedCentral
19.
Zurück zum Zitat Rajala A, Gupta VK, Anderson RE, Rajala RV. Light activation of the insulin receptor regulates mitochondrial hexokinase. A possible mechanism of retinal neuroprotection. Mitochondrion. 2013;13:566–76.CrossRefPubMed Rajala A, Gupta VK, Anderson RE, Rajala RV. Light activation of the insulin receptor regulates mitochondrial hexokinase. A possible mechanism of retinal neuroprotection. Mitochondrion. 2013;13:566–76.CrossRefPubMed
20.
Zurück zum Zitat Wyse Jackson AC, Cotter TG. The synthetic progesterone Norgestrel is neuroprotective in stressed photoreceptor-like cells and retinal explants, mediating its effects via basic fibroblast growth factor, protein kinase A and glycogen synthase kinase 3beta signalling. Eur J Neurosci. 2016;43:899–911.CrossRefPubMed Wyse Jackson AC, Cotter TG. The synthetic progesterone Norgestrel is neuroprotective in stressed photoreceptor-like cells and retinal explants, mediating its effects via basic fibroblast growth factor, protein kinase A and glycogen synthase kinase 3beta signalling. Eur J Neurosci. 2016;43:899–911.CrossRefPubMed
21.
Zurück zum Zitat van Weeren PC, de Bruyn KM, de Vries-Smits AM, van Lint J, Burgering BM. Essential role for protein kinase B (PKB) in insulin-induced glycogen synthase kinase 3 inactivation. Characterization of dominant-negative mutant of PKB. J Biol Chem. 1998;273:13150–6.CrossRefPubMed van Weeren PC, de Bruyn KM, de Vries-Smits AM, van Lint J, Burgering BM. Essential role for protein kinase B (PKB) in insulin-induced glycogen synthase kinase 3 inactivation. Characterization of dominant-negative mutant of PKB. J Biol Chem. 1998;273:13150–6.CrossRefPubMed
22.
Zurück zum Zitat Isiegas C, Marinich-Madzarevich JA, Marchena M, Ruiz JM, Cano MJ, de la Villa P, Hernandez-Sanchez C, de la Rosa EJ, de Pablo F. Intravitreal injection of proinsulin-loaded microspheres delays photoreceptor cell death and vision loss in the rd10 mouse model of retinitis Pigmentosa. Invest Ophthalmol Vis Sci. 2016;57(8):3610.CrossRefPubMed Isiegas C, Marinich-Madzarevich JA, Marchena M, Ruiz JM, Cano MJ, de la Villa P, Hernandez-Sanchez C, de la Rosa EJ, de Pablo F. Intravitreal injection of proinsulin-loaded microspheres delays photoreceptor cell death and vision loss in the rd10 mouse model of retinitis Pigmentosa. Invest Ophthalmol Vis Sci. 2016;57(8):3610.CrossRefPubMed
23.
Zurück zum Zitat Malaguarnera R, Sacco A, Voci C, Pandini G, Vigneri R, Belfiore A. Proinsulin binds with high affinity the insulin receptor isoform a and predominantly activates the mitogenic pathway. Endocrinology. 2012;153:2152–63.CrossRefPubMed Malaguarnera R, Sacco A, Voci C, Pandini G, Vigneri R, Belfiore A. Proinsulin binds with high affinity the insulin receptor isoform a and predominantly activates the mitogenic pathway. Endocrinology. 2012;153:2152–63.CrossRefPubMed
24.
Zurück zum Zitat Fernandez-Sanchez L, Lax P, Isiegas C, Ayuso E, Ruiz JM, de la Villa P, Bosch F, de la Rosa EJ, Cuenca N. Proinsulin slows retinal degeneration and vision loss in the P23H rat model of retinitis pigmentosa. Hum Gene Ther. 2012;23:1290–300.CrossRefPubMedPubMedCentral Fernandez-Sanchez L, Lax P, Isiegas C, Ayuso E, Ruiz JM, de la Villa P, Bosch F, de la Rosa EJ, Cuenca N. Proinsulin slows retinal degeneration and vision loss in the P23H rat model of retinitis pigmentosa. Hum Gene Ther. 2012;23:1290–300.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Ransohoff RM. How neuroinflammation contributes to neurodegeneration. Science. 2016;353:777–83.CrossRefPubMed Ransohoff RM. How neuroinflammation contributes to neurodegeneration. Science. 2016;353:777–83.CrossRefPubMed
26.
Zurück zum Zitat Beurel E, Michalek SM, Jope RS. Innate and adaptive immune responses regulated by glycogen synthase kinase-3 (GSK3). Trends Immunol. 2010;31:24–31.CrossRefPubMed Beurel E, Michalek SM, Jope RS. Innate and adaptive immune responses regulated by glycogen synthase kinase-3 (GSK3). Trends Immunol. 2010;31:24–31.CrossRefPubMed
27.
Zurück zum Zitat Jope RS, Cheng Y, Lowell JA, Worthen RJ, Sitbon YH, Beurel E. Stressed and inflamed, can GSK3 be blamed? Trends Biochem Sci. 2017;42:180–92.CrossRefPubMed Jope RS, Cheng Y, Lowell JA, Worthen RJ, Sitbon YH, Beurel E. Stressed and inflamed, can GSK3 be blamed? Trends Biochem Sci. 2017;42:180–92.CrossRefPubMed
28.
Zurück zum Zitat Barcelona PF, Saragovi HU. A pro-nerve growth factor (proNGF) and NGF binding protein, alpha2-macroglobulin, differentially regulates p75 and TrkA receptors and is relevant to neurodegeneration ex vivo and in vivo. Mol Cell Biol. 2015;35:3396–408.CrossRefPubMedPubMedCentral Barcelona PF, Saragovi HU. A pro-nerve growth factor (proNGF) and NGF binding protein, alpha2-macroglobulin, differentially regulates p75 and TrkA receptors and is relevant to neurodegeneration ex vivo and in vivo. Mol Cell Biol. 2015;35:3396–408.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Shi Z, Rudzinski M, Meerovitch K, Lebrun-Julien F, Birman E, Di Polo A, Saragovi HU. Alpha2-macroglobulin is a mediator of retinal ganglion cell death in glaucoma. J Biol Chem. 2008;283:29156–65.CrossRefPubMedPubMedCentral Shi Z, Rudzinski M, Meerovitch K, Lebrun-Julien F, Birman E, Di Polo A, Saragovi HU. Alpha2-macroglobulin is a mediator of retinal ganglion cell death in glaucoma. J Biol Chem. 2008;283:29156–65.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Platon-Corchado M, Barcelona PF, Jmaeff S, Marchena M, Hernandez-Pinto AM, Hernandez-Sanchez C, Saragovi HU, de la Rosa EJ. p75 NTR antagonists attenuate photoreceptor cell loss in murine models of retinitis pigmentosa. Cell Death Dis. 2017;8:e2922.CrossRefPubMedPubMedCentral Platon-Corchado M, Barcelona PF, Jmaeff S, Marchena M, Hernandez-Pinto AM, Hernandez-Sanchez C, Saragovi HU, de la Rosa EJ. p75 NTR antagonists attenuate photoreceptor cell loss in murine models of retinitis pigmentosa. Cell Death Dis. 2017;8:e2922.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Morales-Garcia JA, Palomo V, Redondo M, Alonso-Gil S, Gil C, Martinez A, Perez-Castillo A. Crosstalk between phosphodiesterase 7 and glycogen synthase kinase-3: two relevant therapeutic targets for neurological disorders. ACS Chem Neurosci. 2014;5:194–204.CrossRefPubMedPubMedCentral Morales-Garcia JA, Palomo V, Redondo M, Alonso-Gil S, Gil C, Martinez A, Perez-Castillo A. Crosstalk between phosphodiesterase 7 and glycogen synthase kinase-3: two relevant therapeutic targets for neurological disorders. ACS Chem Neurosci. 2014;5:194–204.CrossRefPubMedPubMedCentral
Metadaten
Titel
Modulation of GSK-3 provides cellular and functional neuroprotection in the rd10 mouse model of retinitis pigmentosa
verfasst von
Alonso Sánchez-Cruz
Beatriz Villarejo-Zori
Miguel Marchena
Josefa Zaldivar-Díez
Valle Palomo
Carmen Gil
Ignacio Lizasoain
Pedro de la Villa
Ana Martínez
Enrique J. de la Rosa
Catalina Hernández-Sánchez
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Neurodegeneration / Ausgabe 1/2018
Elektronische ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-018-0251-y

Weitere Artikel der Ausgabe 1/2018

Molecular Neurodegeneration 1/2018 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.