Next Article in Journal
About the Transient Effects of Synthetic Amorphous Silica: An In Vitro Study on Macrophages
Next Article in Special Issue
Correlation between the Serum Concentration of Vitamin A and Disease Severity in Patients Carrying p.G90D in RHO, the Most Frequent Gene Associated with Dominant Retinitis Pigmentosa: Implications for Therapy with Vitamin A
Previous Article in Journal
Yeast Mannan-Rich Fraction Modulates Endogenous Reactive Oxygen Species Generation and Antibiotic Sensitivity in Resistant E. coli
Previous Article in Special Issue
Signal Peptide Variants in Inherited Retinal Diseases: A Multi-Institutional Case Series
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia

1
Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
2
Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, AP-HP, 75010 Paris, France
3
Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
4
Wavefront-Engineering Microscopy Group, Neurophotonics Laboratory, CNRS UMR8250, Paris Descartes University, 75270 Paris, France
5
Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4056 Basel, Switzerland
6
Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
7
Zeiss Vision Lab, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
8
Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
9
Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
10
Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France
11
Académie des Sciences, Institut de France, 75006 Paris, France
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 219; https://doi.org/10.3390/ijms24010219
Submission received: 4 October 2022 / Revised: 13 December 2022 / Accepted: 14 December 2022 / Published: 22 December 2022

Abstract

:
Mutations in GPR179 are one of the most common causes of autosomal recessive complete congenital stationary night blindness (cCSNB). This retinal disease is characterized in patients by impaired dim and night vision, associated with other ocular symptoms, including high myopia. cCSNB is caused by a complete loss of signal transmission from photoreceptors to ON-bipolar cells. In this study, we hypothesized that the lack of Gpr179 and the subsequent impaired ON-pathway could lead to myopic features in a mouse model of cCSNB. Using ultra performance liquid chromatography, we show that adult Gpr179−/− mice have a significant decrease in both retinal dopamine and 3,4-dihydroxyphenylacetic acid, compared to Gpr179+/+ mice. This alteration of the dopaminergic system is thought to be correlated with an increased susceptibility to lens-induced myopia but does not affect the natural refractive development. Altogether, our data added a novel myopia model, which could be used to identify therapeutic interventions.

1. Introduction

Myopia is the most common ocular dysfunction worldwide with an alarming rise in prevalence, especially in Southeast Asia [1,2,3,4]. It is characterized by an abnormal increase in the axial length of the eye during refractive development (i.e., emmetropization process), leading to a disturbed far-sight but normal near-sight [1,2,5]. Previous studies of myopic patients and animal models have expanded the knowledge of mechanisms implicated in the onset and progression of myopia, but many cellular and molecular processes are still poorly understood. Myopia can be divided according to the refractive error of a patient between low myopia (≤−0.5 D) and high myopia (≤−6.00 D). High myopia can be a cause of blindness and lead to other ocular diseases such as glaucoma, retinal detachment and cataract [6,7]. Many studies have shown that exposure to outdoor light inhibits myopia onset and progression in both humans [7,8,9,10,11,12,13,14] and experimental animal models [15,16,17,18,19,20]. To date, many models from different species have been proposed to study myopia. Although larger animal models more similar to humans—such as monkeys and tree shrews—would be of interest, technical and ethical issues hinder their development and use. In contrast, the generation of mouse models is manageable but displays pros and cons. On the one hand, both the genetics and environment can be controlled and modified. On the other hand, the small size of the murine eye leads to difficulties in manipulation and interpretation of results during intraocular injection and ocular measurements [21,22,23,24], and to difficulties defining an end point for the emmetropization process. It was shown that one diopter change in refractive error corresponds to an increase of 5.4–6.5 μm in axial length of C57BL/6 mouse eyes [25,26] as opposed to the 280 µm to 400 µm changes in human children and adults, respectively [27,28]. Therefore, in small animals, it is necessary to induce myopia in order to measure detectable refractive errors until a technology overcomes this issue. Myopia induction can be performed using lens-induced myopia (LIM) or form deprivation myopia (FDM) protocols, both of which have been validated in mice and chickens [22,29,30,31,32,33,34].
Although the precise molecular mechanisms of refractive error onset and inhibition remain to be elucidated, previous work has highlighted the impact of the retinal content of the neurotransmitter dopamine (DA) and its receptors upon refractive development and myopia [35,36,37,38,39,40]. Indeed, in the retina, DA is synthesized by dopaminergic amacrine cells (DACs), its secretion is regulated by light conditions [41,42,43,44] and it requires a functional ON-pathway [29,45]. The retinal level of the DA degradation metabolite, 3,4 dihydroxyphenylacetic acid (DOPAC) is a commonly used marker of DA release and the DOPAC/DA ratio is considered a marker of DA turnover. It was shown that DA and DOPAC levels are reduced in a variety of myopia experimental models including FDM and LIM [17,43,44,46,47,48] and experimentally increased retinal DA levels inhibit the development of FDM and LIM in amphibians [42], mammals [43,44] and avian [48,49], and vice versa [38,40].
Several inherited retinal diseases are associated with high myopia (syndromic myopia) [7] including cCSNB [50] where patients display high myopia with a median error of −7.4 D [50,51]. cCSNB is a group of genetically heterogeneous and non-progressive retinal disorders. Its main clinical features consist of an impairment of dim light vision, often associated with other ocular signs such as high myopia, nystagmus and/or strabismus [50,51,52,53,54,55,56]. cCSNB is characterized by a normal a-wave but a severe or complete loss of the b-wave under scotopic conditions and altered b-waves under photopic conditions, as measured with electroretinography. Such abnormal electroretinograms are directly caused by the dysfunction of the ON-BC pathway [50,52,56,57]. To date, mutations causing cCSNB in patients were found in NYX, TRPM1, GRM6, GPR179 and LRIT3 genes [50,58,59,60,61,62,63,64,65,66,67]. Animal models mimicking those mutations already exist and have been extensively characterized: Nyx (also known as nob (no b-wave)) Grm6, Trpm1, Gpr179 or Lrit3 [29,62,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86]. Previous studies have shown that nob mice (carrying a null mutation in Nyx, which causes a loss of function of nyctalopin) display a higher susceptibility to FDM than wild-type littermates. The retinas of ungoggled nob mice also have a lower level of light-induced DOPAC and an overall decrease in both DOPAC and DA levels compared with wild-type mice [29]. Similar results were found in mice lacking Grm6 [45]. Interestingly, a mouse model with cone dysfunction (Gnat2−/−) also displayed a higher susceptibility to FDM, but the levels of retinal DOPAC or DA were unchanged [87]. Despite the fact that all the mutated genes in cCSNBs seem to affect the same pathway, and that patients and all animal models display ON-bipolar cell (ON-BC) transmission defects as shown by electroretinography (for review, see [50]), some differences have been noticed. Lrit3−/− mice exhibit a slight decrease in thickness of the inner nuclear layer [77], while such an observation was absent in retinas of Gpr179−/− mice [74].
All proteins encoded by cCSNB-related genes are involved in synaptic transmission between photoreceptors and ON-BCs [50]. In this study, we decided to focus our experiments on the Gpr179−/− cCSNB mouse model [74]. GPR179 is an orphan G-protein coupled receptor located at the dendritic tips of ON-BCs [75]. It was shown to interact with the presynaptic transmembrane protein pikachurin and the post synaptic RGS11/9 complex to modulate the activation of TRPM1 channel which is the final actor in the mGluR6-mediated cascade leading to the depolarization of ON-BCs [50,82].
In the mammalian retina, detection of light increments and decrements is mediated by the ON and OFF pathways, respectively. In a functional ON pathway, ON-BCs convey the visual information from photoreceptors to ON-retinal ganglion cells (ON-RGCs) in two different ways according to the origin of their input. ON-cone bipolar cells (ON-CBCs) receive direct input from cone photoreceptors and synapse onto ON-RGCs. At low light intensity, rod-bipolar cells (RBCs) receive their input from rods and synapse onto AII amacrine cells (AII-ACs) which, in turn, activate ON-CBCs via gap junctions and inhibit OFF-cone bipolar cells (OFF-CBCs) through inhibitory synapses. ON-CBCs then synapse onto ON-RGCs. In a functional OFF pathway, OFF-CBCs convey the visual information from cone photoreceptors (but also from a minority of rods) to OFF-retinal ganglion cells (OFF-RGCs). Ultimately, RGCs indirectly drive the visual information to the visual cortex through the optic nerve and thalamus [88,89]. In addition to these signaling pathways, the activity of ACs is noteworthy since DACs receive input from AII-ACs and CBCs and some subtypes of most major classes of retinal neurons harbor dopamine receptors (D1R to D4R) [90]. As functional GPR179 is necessary for a proper ON signaling, and as DACs receive indirect excitatory input from ON-BCs, one might expect that the loss of GPR179 could cause a reduced activity of DACs and thus, a reduction in DA release.
The aim of our study was to monitor whether the lack of GPR179 in a novel model of cCSNB [74] has an impact upon DA turnover and induced-myopia. We measured the retinal DA and DOPAC levels, the natural refractive development and the sensitivity to LIM induction in Gpr179−/− mice in order to improve our understanding of the role of cCSNB molecules and the ON signaling cascade upon retinal function and myopia onset and progression.

2. Results

2.1. Measurement of Retinal Levels of DA and DOPAC

To determine whether the loss of Gpr179 induces a change in dopamine metabolism and activity, we quantified the levels of DA and DOPAC in the retinas of adult (post-natal day 80, P80) Gpr179+/+ and Gpr179−/− mice after 4 hours of light adaptation (Figure 1) at 50 lux. Gpr179−/− retinas display a significant decrease in DA (Figure 1A) compared with wild-type littermates. Mean DA levels were 3022 ± 214 fmoles/retina in Gpr179+/+ compared with 1310 ± 135 fmoles/retina in Gpr179−/−. Similarly, Gpr179−/− retinas display a significant decrease in DOPAC (Figure 1B) compared with wild-type littermates. Mean DOPAC levels were 844 ± 38 fmoles/retinas in Gpr179+/+ compared with 413 ± 25 fmoles/retinas. No difference in the DOPAC/DA ratio was observed in Gpr179−/− compared to Gpr179+/+ (Figure 1C). These results indicate that mice lacking Gpr179 exhibit disturbances in dopamine metabolism.

2.2. Kinetics of Refractive Development

In order to establish whether the loss of Gpr179 and the resulting dysfunctional ON-BC pathway could lead to change in natural refractive development, we measured the refractive state of both eyes of Gpr179+/+ and Gpr179−/− mice from 3 to 9 weeks of age (Figure 2). In both genotypes tested, we observed a hyperopic shift from 3- to 6-weeks-old, starting from 0 to +1 D at 3-weeks-old and reaching +3 D to +4 D at 6-weeks-old. From 6- to 9-weeks-old a plateau was reached at +3 to +4 D for both genotypes. Overall, no significant change in the refractive state was observed between Gpr179−/− mice and their wild-type littermates, therefore loss of GPR179 did not induce any measurable change of refractive development.

2.3. Assessment of Sensitivity to Myopia Induction

To test whether the loss of GPR179 could affect the susceptibility to environmentally-induced myopia, we generated a LIM protocol in Gpr179+/+ and Gpr179−/− mice from P21 to P42 and measured the mean interocular shift (called myopic shift when negative) at different time points: P21 (Figure 3; 0-week post-goggling), P35 (2 weeks post-goggling) and P42 (3 weeks post-goggling). At 0-week, no myopic shift was observed in Gpr179+/+ nor in Gpr179−/− mice and no difference of myopic shift between the two genotypes was measured. At 2 weeks post-goggling, we recorded a significant difference in the myopic shift between Gpr179+/+ and Gpr179−/− mice. Myopic shift reached −7.0 ± 0.9 D in Gpr179+/+ and −10.9 ± 0.8 D in Gpr179−/− mice (p < 0.01). At 3 weeks post-goggling, the difference in the myopic shift between Gpr179+/+ and Gpr179−/− mice increased compared to week 2. The myopic shift in Gpr179+/+ mice slightly increased to −8.3 ± 0.6 D while it reached −16.1 ± 0.8 D in Gpr179−/− mice (p < 0.001). Overall, these data demonstrate an impact of the loss of GPR179 upon the sensitivity to lens-induced myopia.

3. Discussion

For the last two decades, the impact of ON-pathway defects upon refractive development and myopia onset has raised attention. Although the detailed mechanisms of normal and myopic refractive development are still unclear, the development of new induction protocols and the extensive use of transgenic models, that allow for the manipulation of both environmental and genetic factors, help resolve the signaling cascades involved. To date, many factors have been proposed to trigger or stop the growth of the eyeball and thus, to impact refractive development [39,43,91,92,93,94,95,96]. Among them, the dopaminergic system is the most characterized [17,47,90,97]. In this study, we report for the first time the presence of myopic features in the Gpr179 mouse model [74].
Our experiments revealed drastic decreases of both DA and DOPAC levels in the light-adapted retinas of Gpr179−/− mice compared to their wild-type littermates (Figure 1). DA is reduced in Gpr179−/− mice by two-thirds while DOPAC is halved. Functional GPR179 is necessary for a normal ON signaling, and DACs receive indirect excitatory input from ON-BCs. Furthermore, stimulation of the ON pathway induces the release of DA via the activation of DACs [42,90,98]. Thus, it has been hypothesized that defects in ON-BCs signaling could impact DA release [29,45,99]. Our results are in line with previous studies addressing this hypothesis [29,45] Still, some differences with previous findings are noteworthy. Here, both DA and DOPAC levels are reduced in Gpr179−/− mice while only DOPAC was found to be decreased in Grm6−/− mouse retinas [45]. In the retinas of nob mice, both DA and DOPAC levels were lower than in wild-type mice [29]. A disparity in tested ages was proposed to explain some of the observed differences as an impact of age upon retinal DA and DOPAC levels has been reported [100]. Our results are consistent with this explanation since both Gpr179−/− and nob [70] mice were 12 weeks old but mice lacking Grm6 [45] were 16 weeks old. Another explanation could involve the lighting conditions used for light adaptation [17] which may have been different in the previous studies [29,45] compared to ours. A more interesting explanation might be that although both GRM6 and GPR179 are necessary for ON-BC signaling and are thought to be expressed in all ON-BCs and similarly localized to their dendritic tips, it is not known if the physiological consequences of their absence are the same. Indeed, the light responses of retinal ganglion cells (downstream of ON-BCs) are different in mice with different Grm6 mutant alleles [83], thus it is possible that DACs are differentially affected in Grm6−/− and Gpr179−/− mice.
Our light adaptation was mesopic. Thus, one can hypothesize that residual ON or OFF activation could affect DA metabolism since DACs receive inputs from ON and, to a lesser extent, from OFF pathways [90]. This hypothesis seems unlikely as (1) all the cCSNB mouse lines tested display a similar loss of ON-pathway function [70,74,86]. (2) To our knowledge OFF responses have not been characterized in the Gpr179−/− mouse line, but other studies show that the OFF pathway is globally unaltered in cCSNB patients [50,101] and mouse models [77,102]. (3) Experiments performed in a Vsx1−/− mouse, a model with dysfunctional OFF bipolar cells [103], revealed no impact of the OFF pathway defect upon refractive development and myopia onset [104] nor on dopamine metabolism in adult Vsx1−/− retinas [99].
The DOPAC/DA ratio is unchanged in Gpr179−/− compared to Gpr179+/+ (Figure 1C). Several studies have noted the impact of an altered DOPAC/DA ratio upon sensitivity to myopia induction [29,43,45,105]. In our study, both DOPAC and DA levels are reduced in Gpr179−/− mice by similar levels, leading to an unchanged DOPAC/DA ratio. Overall, our results are in line with the hypothesis of a defective ON-pathway causing a reduction of retinal dopamine.
Strikingly, comparing the natural refractive development of Gpr179+/+ and Gpr179−/− mice, we found no difference in the refractive state between the two genotypes across age. Previous studies performed on other cCSNB models display variable results: Nyx−/− mice become more hyperopic than wild-type littermates [29], while Grm6−/− mice display a more myopic development [45]. Furthermore, both Gpr179+/+ and Gpr179−/− mice are more myopic in general than some genetic mouse models [29,30,45], but similar to others [17,104,106,107]. Both genetic [104,108] and environmental [17,107] factors can influence refractive development. We used the same mouse background strain, C57BL/6J as others [29,45,104,108], but as mentioned above, lighting conditions can differ between laboratories. One study also found an impact of circadian rhythm upon axial length [109], with a range of 20 µm difference between the maximum and the minimum, which would cause a theoretical change in refractive state of about 4–5 D. In the present study, we did not consider this potential cause of variability. Another cause of disparity could be the use of eye drops and the absence of anesthetics in our experimental paradigm. It was shown that intra-peritoneal injection of ketamine/xylazine could induce a hyperopic shift in mice [110]. We tested the use of isoflurane inhalation instead but found the same issue as with ketamine/xylazine anesthesia (data not shown). However, the time course of refractive development follows the same pattern as in other studies: a hyperopic shift followed by a plateau reached at 6 weeks of age [29,45,104]. Many studies have noted the difficulty of measuring myopia in small animals because of the small eye artifact [29,45,111]. Our data confirm that measurement of myopia in small animals often requires an induction protocol.
By developing a 3-week-long lens-induced myopia protocol, we found a substantial increase of the myopic shift in Gpr179−/− mice compared to wild-type littermates (Figure 3). Consistent with previous findings with cCSNB models, this myopic shift is significant at 2 weeks post-goggling [29,45], but continues to increase until 3 weeks. Surprisingly, this myopic shift is also much larger in Gpr179+/+ mice than in the control mice used in those previous studies. As mentioned before, we used the same mouse strain, but the lightning environment might have had an impact. However, the most relevant cause of difference could be found in the induction protocol. We used a −25 D lens as opposed to monocular deprivation and started the induction one week earlier than in the studies mentioned above. Our results are in line with previous studies from other groups showing that starting the induction at P21 [46] or P24 [112] instead of P28 causes a more substantial increase in myopic shift.
We need to elucidate whether the myopic shift induced by our LIM protocol directly leads to a decrease in retinal levels of DA and/or DOPAC in wild-type and in Gpr179−/− mice. Interestingly, recent studies showed that injecting DA agonists suppresses FDM in several animal models [48,113,114]. In addition, induction of FDM seemed to decrease retinal levels of DA in guinea pigs [115], chickens [116] and primates [91] but not in wild-type mice [117]. This may be at least partly explained by the fact that most mice used in myopia studies are melatonin-deficient mice (i.e., C57BL/6J mice). Melatonin is released by photoreceptors, is implicated in circadian rhythm, and seems to inhibit dopamine release. Dopamine and melatonin act in opposite ways, interacting with each other to form an inhibitory feedback loop [118]. In support of the notion that melatonin is required for FDM-related retinal DA changes, a recent study showed that melatonin-proficient mice (i.e., CBA/CaJ) display reduced retinal levels of DA during FDM [119]. Similarly, this may explain a part of the residual dopamine release in the retinas of cCSNB mouse models observed in our study and by others [29,45]. Further experiments are required to determine whether the causal link between DA metabolism and sensitivity to myopia induction is unidirectional or bidirectional in mice. Mechanisms implicated in LIM-mediated myopic shift seem to be less dependent on DA metabolism than those mediated by FDM [43, 47], but are clearly not independent either. This can be explained by the loss of the timing cue of lighting occurring during FDM but not during LIM [47].
It is noteworthy that injecting L-AP4 in chicken eyes causes a hyperopic shift in ungoggled eyes but does not have an impact on form-deprived ones [120,121]. L-AP4 is an agonist of group III metabotropic glutamate receptors. In the retina, it stimulates mGluR6 and whether it causes the ON-BCs to be constitutively hyperpolarized remains unknown. To our knowledge, there is still little evidence on the resting state of ON-BCs in cCSNB models. Whole-cell patch-clamp recordings of RBCs from nob mice retinal slices revealed small holding currents, indicating that channels were mostly closed or absent [122]. Further studies focusing on the differential effect of L-AP4 upon ungoggled eyes and form-deprived ones might be of interest to decipher the mechanisms leading to myopia in cCSNBs.
Altogether, our results indicate that the loss of GPR179 induces disturbances in the dopaminergic system. The ON-pathway defect due to the lack of GPR179 does not have any measurable impact upon refractive development but increases the sensitivity to LIM. Overall, our data strengthen the hypothesis that ON-pathway deficiency can be associated with myopia onset through alteration of dopaminergic signaling. This work validates the potential use of cCSNB models to study myopia. Thus, we think they can be used to identify new genes related to myopia in cCSNB. For instance, by performing RNAseq studies upon cCSNB models and patients, we may find genes implicated in syndromic and/or common myopia.

4. Material and Methods

4.1. Animal Care and Ethical Statement

All animal procedures were performed according to the Council Directive 2010/63EU of the European Parliament and the Council of 22 September 2010, on the protection of animals used for scientific purposes, with the National Institutes of Health guidelines and with the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research. They were approved by the French Minister of National Education, Superior Education and Research. Mouse lines and projects were registered as following: refractometry: APAFIS #27474 2020100110251857 v5. Mice were kept in 12:12 hour light:dark cycles with mouse chow and water ad libitum.

4.2. Polymerase Chain Reaction (PCR) Genotyping

DNA was extracted from mouse tails with 50 mM NaOH after incubation at 95 °C for 30 min. Wild-type and mutant allele were amplified independently using a polymerase (HOT FIREPol, Solis Biodyne, Tartu, Estonia), a common forward primer: mGpr179_EF (5′-CTGCCCCCACAGAATGTTCCCA-3′) and two specific reverse primers: mGpr179_Er2 (5′-CACCGCCTCTTTACTCTGCCCA-3′) for the wild-type allele and mGpr179_Kr (5′-GGGCAAGAACATAAAGTGACCCTCC-3′) for the mutant allele. The following PCR program was used: 15 min at 95 °C for denaturation, 35 cycles of 45 sec at 95 °C, 1 min at 60 °C, and 1.3 min at 72 °C. A final extension for 10 min at 72 °C was performed. This generates the following amplicons: PCR using mGpr179_EF and mGpr179_Er2 primers amplifies a product of 146 base pairs (bp) for the wild-type allele and no product for the mutant allele, PCR using mGpr179_EF and mGpr179_Kr primers amplifies no product for the wild-type allele and a 303 bp product for the mutant allele. PCR products were separated by electrophoresis on 2% agarose gels, stained with ethidium bromide, and visualized using a documentation system (Molecular Imager® Gel DocTM XR+ System, Bio-Rad, Hercules, CA, USA).

4.3. DA and DOPAC Measurements

After 4 hours of light adaptation in a light controlled room at 50 lux, mice were sacrificed at 12 pm by CO2 inhalation followed by cervical dislocation. Retinas were isolated, frozen in liquid nitrogen and stored at −80 °C. Amounts of DA and DOPAC were quantified with ultra performance liquid chromatography (UPLC) with coulometric detection adapted from [123]. The UPLC equipment consisted of a system (Ultimate 3000, Thermo Fisher Waltham, MA, USA) in isocartic mode with two working electrodes (Coulochem III, Thermo Fisher, Waltham, MA, USA). Analytes were separated on a C18 column (150 mm × 4.6 mm, 3 µm particle size, BDS Hypersil, Thermo Fisher, Waltham, MA, USA), maintained at 40 °C, with a mobile phase (Thermo Fisher Test). Briefly, tissue samples were crushed for 30 s (Ultraturax, IKA, Staufen, Germany) in 0.2 M perchloric acid containing EDTA and ascorbic acid (0.2 µM each). After centrifugation at 25,000x g for 30 min at 4 °C, the supernatants were passed through a 10 kD cutoff filter (Nanosep, Port Washington, NY, USA) at 12,000 g, for 10 mins at 4 °C. Finally, 50 µL of the ultrafiltrates were injected. The two serial coulometric electrodes with applied potentials −100 and 350 mV were used for the measurements of DA and DOPAC.
Statistical analyses were performed using Prism 9.1.2, GraphPad (GraphPad v7 Software Inc., La Jolla, CA, USA). Statistical significance was measured with a non-parametric two-tailed Mann–Whitney test.

4.4. Lens-Induced Myopia

To induce myopia, P21 mice were anesthetized by isoflurane inhalation (5% induction, 2% maintenance). The scalp was cut to expose 1.2 cm of the skull in the rostrocaudal axis. Two intracranial screws were implanted on both left and right sides of the skull at y = −2 mm from the bregma. A homemade goggle frame was placed on the skull and fixed using dental cement (FujiCEM™, Phymep© Cat #900903, Paris, France). This goggle frame, adapted from X. Jiang et al.; 2018 [46], was built in resin using a 3D printer. The −25 D lenses were stuck on the frame using glue (vetbond™, Phymep©, Paris, France). Stitches were used to avoid displacement of lens by mice. The −25 D lens was always placed in front of the right eye.

4.5. Refractometry

After a dark adaptation for 30 min, eye drops were used to dilate the pupils: 0.5% mydriaticum© (Théa, Clermont Ferrand, France), 5% neosynephrine (Europhta, Monaco). The mice were placed on a homemade restraining platform in front of an eccentric infrared photorefractometer calibrated as described previously [32]. Calibration was verified using lenses of increasing power, from −10 D to +10 D placed in front of a mouse eye. The mouse was positioned so that the first Purkinje image was in the center of the pupil. The data were then recorded using software designed by F. Schaeffel [32]. We collected two data sets per animal, one per eye, each data set consisted of 90 measures, each of which was the mean of 10 successful measurements (in total: 1800 points/animal). For refractive development experiments, the measurements were performed once per week, every week from 3- to 9-weeks-old and the mean refractive state of both eyes was used for statistical analysis. To evaluate the sensitivity to myopia induction, the difference between goggled and ungoggled eye (referred to as interocular shift) measured at post-natal day 21 (P21, day of surgical procedure), P35 (14 days after surgery) and P42 (21 days after surgery) was used for statistical analysis. Only the mice displaying less than 2 D of interocular shift at P21 were used for the induction protocol. Statistical analyses were performed using Prism 9.1.2, GraphPad (GraphPad v7 Software Inc., La Jolla, CA, USA). Statistical significance was measured with a two-way ANOVA test.

Author Contributions

Conceptualization, C.Z. and S.P.; methodology, validation, investigations, formal analysis, data curation, B.W., J.C., R.D., C.T., C.M., H.F. and R.G.; resources, C.Z., I.A., O.M., S.P. and J.A.S.; writing—original draft preparation, B.W.; writing—review and editing, B.W., F.S., I.A. and C.Z.; supervision, C.Z. and I.A.; funding acquisition, C.Z., S.P., I.A. and J.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Agence Nationale de la Recherche (ANR-12-BSVS1–001201_GPR179) and (ANR-22-CHIN-0006) (CZ); Institut Clinique de la Souris-ICS, Illkirch, France (partial financial support to create the mouse model (CZ)); Retina France; Valentin Haüy and AFM (CZ), IRP-INSERM (CZ and RD); Prix Dalloz pour la recherche en ophtalmologie and Fondation Dalloz—Institut de France (CZ); Fondation Voir et Entendre (CZ); Fondation de l’œil—Fondation de France (IA), Ville de Paris and Region Ile de France; Labex Lifesenses (reference ANR-10-LABX-65), supported by French state funds managed by the ANR within the Investissements d’Avenir programme (ANR-11-IDEX-0004-0); the Programme Investissements d’Avenir IHU FOReSIGHT (ANR-18-IAHU-01); National Institutes of Health grant EY029985 (RD) and Essilor (BW).

Institutional Review Board Statement

All animal procedures were performed according to the Council Directive 2010/63EU of the European Parliament and the Council of 22 September 2010 regarding the protection of animals used for scientific purposes, and according to the National Institutes of Health guidelines and the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research. The project was approved by the French Minister of National Education, Superior Education and Research (authorization delivered on the 30 December 2020, APAFIS #27474 2020100110251857 v5.

Informed Consent Statement

Non-applicable.

Data Availability Statement

Non-applicable.

Acknowledgments

The authors are grateful to Manuel Simonutti, Quenol Cesar, Julie Dégardin for their valuable help in phenotyping (platform at the Institut de la Vision) and to Mathilde Lappe, Cyril Duchene and Amandine Chocat for animal care (platform at the Institut de la Vision). The authors thank Andrei Tkatchenko for advice regarding set-up of the restraining platform.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wu, P.C.; Huang, H.M.; Yu, H.J.; Fang, P.C.; Chen, C.T. Epidemiology of Myopia. Asia Pac. J. Ophthalmol. 2016, 5, 386–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Cooper, J.; Tkatchenko, A.V. A Review of Current Concepts of the Etiology and Treatment of Myopia. Eye Contact Lens 2018, 44, 231–247. [Google Scholar] [CrossRef] [PubMed]
  3. Holden, B.A.; Fricke, T.R.; Wilson, D.A.; Jong, M.; Naidoo, K.S.; Sankaridurg, P.; Wong, T.Y.; Naduvilath, T.J.; Resnikoff, S. Global Prevalence of Myopia and High Myopia and Temporal Trends from 2000 through 2050. Ophthalmology 2016, 123, 1036–1042. [Google Scholar] [CrossRef] [Green Version]
  4. Dolgin, E. The myopia boom. Nat. News 2015, 519, 276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Flitcroft, D.I.; He, M.; Jonas, J.B.; Jong, M.; Naidoo, K.; Ohno-Matsui, K.; Rahi, J.; Resnikoff, S.; Vitale, S.; Yannuzzi, L. IMI–Defining and Classifying Myopia: A Proposed Set of Standards for Clinical and Epidemiologic Studies. Investig. Ophthalmol. Vis. Sci. 2019, 60, M20–M30. [Google Scholar] [CrossRef] [Green Version]
  6. Ikuno, Y. Overview of the Complications of High Myopia. Retina 2017, 37, 2347–2351. [Google Scholar] [CrossRef]
  7. Bremond-Gignac, D. Myopia in children. Med. Sci. 2020, 36, 763–768. [Google Scholar]
  8. Wu, P.C.; Chen, C.T.; Lin, K.K.; Sun, C.C.; Kuo, C.N.; Huang, H.M.; Poon, Y.C.; Yang, M.L.; Chen, C.Y.; Huang, J.C.; et al. Myopia Prevention and Outdoor Light Intensity in a School-Based Cluster Randomized Trial. Ophthalmology 2018, 125, 1239–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Wu, P.C.; Tsai, C.L.; Wu, H.L.; Yang, Y.H.; Kuo, H.K. Outdoor activity during class recess reduces myopia onset and progression in school children. Ophthalmology 2013, 120, 1080–1085. [Google Scholar] [CrossRef]
  10. Jones, L.A.; Sinnott, L.T.; Mutti, D.O.; Mitchell, G.L.; Moeschberger, M.L.; Zadnik, K. Parental history of myopia, sports and outdoor activities, and future myopia. Investig. Ophthalmol. Vis. Sci. 2007, 48, 3524–3532. [Google Scholar] [CrossRef] [Green Version]
  11. Jones-Jordan, L.A.; Sinnott, L.T.; Chu, R.H.; Cotter, S.A.; Kleinstein, R.N.; Manny, R.E.; Mutti, D.O.; Twelker, J.D.; Zadnik, K. Myopia Progression as a Function of Sex, Age, and Ethnicity. Investig. Ophthalmol. Vis. Sci. 2021, 62, 36. [Google Scholar] [CrossRef]
  12. Mutti, D.O.; Mitchell, G.L.; Moeschberger, M.L.; Jones, L.A.; Zadnik, K. Parental myopia, near work, school achievement, and children’s refractive error. Investig. Ophthalmol. Vis. Sci. 2002, 43, 3633–3640. [Google Scholar]
  13. French, A.N.; Ashby, R.S.; Morgan, I.G.; Rose, K.A. Time outdoors and the prevention of myopia. Exp. Eye Res. 2013, 114, 58–68. [Google Scholar] [CrossRef] [Green Version]
  14. Rose, K.A.; Morgan, I.G.; Ip, J.; Kifley, A.; Huynh, S.; Smith, W.; Mitchell, P. Outdoor activity reduces the prevalence of myopia in children. Ophthalmology 2008, 115, 1279–1285. [Google Scholar] [CrossRef]
  15. Ashby, R.; Ohlendorf, A.; Schaeffel, F. The effect of ambient illuminance on the development of deprivation myopia in chicks. Investig. Ophthalmol. Vis. Sci. 2009, 50, 5348–5354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Smith, E.L., 3rd; Hung, L.F.; Huang, J. Protective effects of high ambient lighting on the development of form-deprivation myopia in rhesus monkeys. Investig. Ophthalmol. Vis. Sci. 2012, 53, 421–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Landis, E.G.; Park, H.N.; Chrenek, M.; He, L.; Sidhu, C.; Chakraborty, R.; Strickland, R.; Iuvone, P.M.; Pardue, M.T. Ambient Light Regulates Retinal Dopamine Signaling and Myopia Susceptibility. Investig. Ophthalmol. Vis. Sci. 2021, 62, 28. [Google Scholar] [CrossRef] [PubMed]
  18. Norton, T.T.; Amedo, A.O.; Siegwart, J.T., Jr. Darkness causes myopia in visually experienced tree shrews. Investig. Ophthalmol. Vis. Sci. 2006, 47, 4700–4707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Li, W.; Lan, W.; Yang, S.; Liao, Y.; Xu, Q.; Lin, L.; Yang, Z. The effect of spectral property and intensity of light on natural refractive development and compensation to negative lenses in guinea pigs. Investig. Ophthalmol. Vis. Sci. 2014, 55, 6324–6332. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, Y.; Ding, H.; Stell, W.K.; Liu, L.; Li, S.; Liu, H.; Zhong, X. Exposure to sunlight reduces the risk of myopia in rhesus monkeys. PLoS ONE 2015, 10, e0127863. [Google Scholar] [CrossRef] [Green Version]
  21. Song, Y.; Zhang, F.; Zhao, Y.; Sun, M.; Tao, J.; Liang, Y.; Ma, L.; Yu, Y.; Wang, J.; Hao, J. Enlargement of the Axial Length and Altered Ultrastructural Features of the Sclera in a Mutant Lumican Transgenic Mouse Model. PLoS ONE 2016, 11, e0163165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Pardue, M.T.; Stone, R.A.; Iuvone, P.M. Investigating mechanisms of myopia in mice. Exp. Eye Res. 2013, 114, 96–105. [Google Scholar] [CrossRef] [PubMed]
  23. Park, H.; Qazi, Y.; Tan, C.; Jabbar, S.B.; Cao, Y.; Schmid, G.; Pardue, M.T. Assessment of axial length measurements in mouse eyes. Optom. Vis. Sci. 2012, 89, 296–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Benavente-Perez, A.; Nour, A.; Troilo, D. Axial eye growth and refractive error development can be modified by exposing the peripheral retina to relative myopic or hyperopic defocus. Investig. Ophthalmol. Vis. Sci. 2014, 55, 6765–6773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Schmucker, C.; Schaeffel, F. In vivo biometry in the mouse eye with low coherence interferometry. Vis. Res. 2004, 44, 2445–2456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Szczerkowska, K.I.; Petrezselyova, S.; Lindovsky, J.; Palkova, M.; Dvorak, J.; Makovicky, P.; Fang, M.; Jiang, C.; Chen, L.; Shi, M.; et al. Myopia disease mouse models: A missense point mutation (S673G) and a protein-truncating mutation of the Zfp644 mimic human disease phenotype. Cell Biosci. 2019, 9, 21. [Google Scholar] [CrossRef] [Green Version]
  27. Atchison, D.A.; Jones, C.E.; Schmid, K.L.; Pritchard, N.; Pope, J.M.; Strugnell, W.E.; Riley, R.A. Eye shape in emmetropia and myopia. Investig. Ophthalmol. Vis. Sci. 2004, 45, 3380–3386. [Google Scholar] [CrossRef] [Green Version]
  28. Cruickshank, F.E.; Logan, N.S. Optical ‘dampening’ of the refractive error to axial length ratio: Implications for outcome measures in myopia control studies. Ophthalmic Physiol. Opt. J. Br. Coll. Ophthalmic Opt. 2018, 38, 290–297. [Google Scholar] [CrossRef] [Green Version]
  29. Pardue, M.T.; Faulkner, A.E.; Fernandes, A.; Yin, H.; Schaeffel, F.; Williams, R.W.; Pozdeyev, N.; Iuvone, P.M. High susceptibility to experimental myopia in a mouse model with a retinal on pathway defect. Investig. Ophthalmol. Vis. Sci. 2008, 49, 706–712. [Google Scholar] [CrossRef]
  30. Park, H.; Tan, C.C.; Faulkner, A.; Jabbar, S.B.; Schmid, G.; Abey, J.; Iuvone, P.M.; Pardue, M.T. Retinal degeneration increases susceptibility to myopia in mice. Mol. Vis. 2013, 19, 2068–2079. [Google Scholar]
  31. Chakraborty, R.; Lacy, K.D.; Tan, C.C.; Park, H.N.; Pardue, M.T. Refractive index measurement of the mouse crystalline lens using optical coherence tomography. Exp. Eye Res. 2014, 125, 62–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Schaeffel, F.; Burkhardt, E.; Howland, H.C.; Williams, R.W. Measurement of refractive state and deprivation myopia in two strains of mice. Optom. Vis. Sci. 2004, 81, 99–110. [Google Scholar] [CrossRef]
  33. Tejedor, J.; de la Villa, P. Refractive changes induced by form deprivation in the mouse eye. Investig. Ophthalmol. Vis. Sci. 2003, 44, 32–36. [Google Scholar] [CrossRef] [PubMed]
  34. Tkatchenko, A.V.; Tkatchenko, T.V.; Guggenheim, J.A.; Verhoeven, V.J.; Hysi, P.G.; Wojciechowski, R.; Singh, P.K.; Kumar, A.; Thinakaran, G.; Williams, C.; et al. APLP2 Regulates Refractive Error and Myopia Development in Mice and Humans. PLoS Genet 2015, 11, e1005432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Chen, S.; Zhi, Z.; Ruan, Q.; Liu, Q.; Li, F.; Wan, F.; Reinach, P.S.; Chen, J.; Qu, J.; Zhou, X. Bright Light Suppresses Form-Deprivation Myopia Development With Activation of Dopamine D1 Receptor Signaling in the ON Pathway in Retina. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2306–2316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Cohen, Y.; Peleg, E.; Belkin, M.; Polat, U.; Solomon, A.S. Ambient illuminance, retinal dopamine release and refractive development in chicks. Exp. Eye Res. 2012, 103, 33–40. [Google Scholar] [CrossRef] [PubMed]
  37. Li, X.X.; Schaeffel, F.; Kohler, K.; Zrenner, E. Dose-dependent effects of 6-hydroxy dopamine on deprivation myopia, electroretinograms, and dopaminergic amacrine cells in chickens. Vis. Neurosci. 1992, 9, 483–492. [Google Scholar] [CrossRef]
  38. Wu, X.H.; Qian, K.W.; Xu, G.Z.; Li, Y.Y.; Ma, Y.Y.; Huang, F.; Wang, Y.Q.; Zhou, X.; Qu, J.; Yang, X.L.; et al. The Role of Retinal Dopamine in C57BL/6 Mouse Refractive Development as Revealed by Intravitreal Administration of 6-Hydroxydopamine. Investig. Ophthalmol. Vis. Sci. 2016, 57, 5393–5404. [Google Scholar] [CrossRef]
  39. Stone, R.A.; Lin, T.; Laties, A.M.; Iuvone, P.M. Retinal dopamine and form-deprivation myopia. Proc. Natl. Acad. Sci. USA 1989, 86, 704–706. [Google Scholar] [CrossRef] [Green Version]
  40. Nickla, D.L.; Totonelly, K. Dopamine antagonists and brief vision distinguish lens-induced- and form-deprivation-induced myopia. Exp. Eye Res. 2011, 93, 782–785. [Google Scholar] [CrossRef] [Green Version]
  41. Hirasawa, H.; Contini, M.; Raviola, E. Extrasynaptic release of GABA and dopamine by retinal dopaminergic neurons. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2015, 370, 20140186. [Google Scholar] [CrossRef] [Green Version]
  42. Boatright, J.H.; Hoel, M.J.; Iuvone, P.M. Stimulation of endogenous dopamine release and metabolism in amphibian retina by light- and K+-evoked depolarization. Brain Res. 1989, 482, 164–168. [Google Scholar] [CrossRef] [PubMed]
  43. Feldkaemper, M.; Schaeffel, F. An updated view on the role of dopamine in myopia. Exp. Eye Res. 2013, 114, 106–119. [Google Scholar] [CrossRef] [PubMed]
  44. Zhou, X.; Pardue, M.T.; Iuvone, P.M.; Qu, J. Dopamine signaling and myopia development: What are the key challenges. Prog. Retin. Eye Res. 2017, 61, 60–71. [Google Scholar] [CrossRef] [PubMed]
  45. Chakraborty, R.; Park, H.N.; Hanif, A.M.; Sidhu, C.S.; Iuvone, P.M.; Pardue, M.T. ON pathway mutations increase susceptibility to form-deprivation myopia. Exp. Eye Res. 2015, 137, 79–83. [Google Scholar] [CrossRef] [Green Version]
  46. Jiang, X.; Kurihara, T.; Kunimi, H.; Miyauchi, M.; Ikeda, S.-I.; Mori, K.; Tsubota, K.; Torii, H.; Tsubota, K. A highly efficient murine model of experimental myopia. Sci. Rep. 2018, 8, 2026. [Google Scholar] [CrossRef] [Green Version]
  47. Morgan, I.G.; Ashby, R.S.; Nickla, D.L.; Guggenheim, J.A. Form deprivation and lens-induced myopia: Are they different? Ophthalmic Physiol. Opt. J. Br. Coll. Ophthalmic Opt. 2013, 33, 355–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Thomson, K.; Karouta, C.; Ashby, R. Form-Deprivation and Lens-Induced Myopia Are Similarly Affected by Pharmacological Manipulation of the Dopaminergic System in Chicks. Investig. Ophthalmol. Vis. Sci. 2020, 61, 4. [Google Scholar] [CrossRef] [PubMed]
  49. Thomson, K.; Morgan, I.; Karouta, C.; Ashby, R. Levodopa inhibits the development of lens-induced myopia in chicks. Sci. Rep. 2020, 10, 13242. [Google Scholar] [CrossRef] [PubMed]
  50. Zeitz, C.; Robson, A.G.; Audo, I. Congenital stationary night blindness: An analysis and update of genotype–phenotype correlations and pathogenic mechanisms. Prog. Retin. Eye Res. 2015, 45, 58–110. [Google Scholar] [CrossRef]
  51. Bijveld, M.M.; Florijn, R.J.; Bergen, A.A.; van den Born, L.I.; Kamermans, M.; Prick, L.; Riemslag, F.C.; van Schooneveld, M.J.; Kappers, A.M.; van Genderen, M.M. Genotype and phenotype of 101 dutch patients with congenital stationary night blindness. Ophthalmology 2013, 120, 2072–2081. [Google Scholar] [CrossRef]
  52. Zeitz, C. Molecular genetics and protein function involved in nocturnal vision. Expert Rev. Ophthalmol. 2007, 2, 467–485. [Google Scholar] [CrossRef]
  53. Dry, K.L.; Van Dorp, D.B.; Aldred, M.A.; Brown, J.; Hardwick, L.J.; Wright, A.F. Linkage analysis in a family with complete type congenital stationary night blindness with and without myopia. Clin. Genet. 1993, 43, 250–254. [Google Scholar] [CrossRef]
  54. Zeitz, C.; Labs, S.; Lorenz, B.; Forster, U.; Uksti, J.; Kroes, H.Y.; De Baere, E.; Leroy, B.P.; Cremers, F.P.; Wittmer, M.; et al. Genotyping microarray for CSNB-associated genes. Investig. Ophthalmol. Vis. Sci. 2009, 50, 5919–5926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Miyake, Y.; Yagasaki, K.; Horiguchi, M.; Kawase, Y.; Kanda, T. Congenital stationary night blindness with negative electroretinogram. A new classification. Arch. Ophthalmol. 1986, 104, 1013–1020. [Google Scholar] [CrossRef] [PubMed]
  56. Bijveld, M.M.; van Genderen, M.M.; Hoeben, F.P.; Katzin, A.A.; van Nispen, R.M.; Riemslag, F.C.; Kappers, A.M. Assessment of night vision problems in patients with congenital stationary night blindness. PLoS ONE 2013, 8, e62927. [Google Scholar] [CrossRef] [Green Version]
  57. Audo, I.; Robson, A.G.; Holder, G.E.; Moore, A.T. The negative ERG: Clinical phenotypes and disease mechanisms of inner retinal dysfunction. Surv. Ophthalmol. 2008, 53, 16–40. [Google Scholar] [CrossRef] [PubMed]
  58. Audo, I.; Bujakowska, K.; Orhan, E.; Poloschek, C.M.; Defoort-Dhellemmes, S.; Drumare, I.; Kohl, S.; Luu, T.D.; Lecompte, O.; Zrenner, E.; et al. Whole-exome sequencing identifies mutations in GPR179 leading to autosomal-recessive complete congenital stationary night blindness. Am. J. Hum. Genet. 2012, 90, 321–330. [Google Scholar] [CrossRef] [Green Version]
  59. Audo, I.; Kohl, S.; Leroy, B.P.; Munier, F.L.; Guillonneau, X.; Mohand-Saïd, S.; Bujakowska, K.; Nandrot, E.F.; Lorenz, B.; Preising, M.; et al. TRPM1 is mutated in patients with autosomal-recessive complete congenital stationary night blindness. Am. J. Hum. Genet. 2009, 85, 720–729. [Google Scholar] [CrossRef] [Green Version]
  60. Zeitz, C.; Jacobson, S.G.; Hamel, C.P.; Bujakowska, K.; Neuillé, M.; Orhan, E.; Zanlonghi, X.; Lancelot, M.E.; Michiels, C.; Schwartz, S.B.; et al. Whole-exome sequencing identifies LRIT3 mutations as a cause of autosomal-recessive complete congenital stationary night blindness. Am. J. Hum. Genet. 2013, 92, 67–75. [Google Scholar] [CrossRef] [Green Version]
  61. Bech-Hansen, N.T.; Naylor, M.J.; Maybaum, T.A.; Sparkes, R.L.; Koop, B.; Birch, D.G.; Bergen, A.A.; Prinsen, C.F.; Polomeno, R.C.; Gal, A.; et al. Mutations in NYX, encoding the leucine-rich proteoglycan nyctalopin, cause X-linked complete congenital stationary night blindness. Nat. Genet. 2000, 26, 319–323. [Google Scholar] [CrossRef]
  62. Peachey, N.S.; Ray, T.A.; Florijn, R.; Rowe, L.B.; Sjoerdsma, T.; Contreras-Alcantara, S.; Baba, K.; Tosini, G.; Pozdeyev, N.; Iuvone, P.M.; et al. GPR179 is required for depolarizing bipolar cell function and is mutated in autosomal-recessive complete congenital stationary night blindness. Am. J. Hum. Genet. 2012, 90, 331–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Li, Z.; Sergouniotis, P.I.; Michaelides, M.; Mackay, D.S.; Wright, G.A.; Devery, S.; Moore, A.T.; Holder, G.E.; Robson, A.G.; Webster, A.R. Recessive mutations of the gene TRPM1 abrogate ON bipolar cell function and cause complete congenital stationary night blindness in humans. Am. J. Hum. Genet. 2009, 85, 711–719. [Google Scholar] [CrossRef]
  64. Van Genderen, M.M.; Bijveld, M.M.; Claassen, Y.B.; Florijn, R.J.; Pearring, J.N.; Meire, F.M.; McCall, M.A.; Riemslag, F.C.; Gregg, R.G.; Bergen, A.A.; et al. Mutations in TRPM1 are a common cause of complete congenital stationary night blindness. Am. J. Hum. Genet. 2009, 85, 730–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Dryja, T.P.; McGee, T.L.; Berson, E.L.; Fishman, G.A.; Sandberg, M.A.; Alexander, K.R.; Derlacki, D.J.; Rajagopalan, A.S. Night blindness and abnormal cone electroretinogram ON responses in patients with mutations in the GRM6 gene encoding mGluR6. Proc. Natl. Acad. Sci. USA 2005, 102, 4884–4889. [Google Scholar] [CrossRef] [Green Version]
  66. Pusch, C.M.; Zeitz, C.; Brandau, O.; Pesch, K.; Achatz, H.; Feil, S.; Scharfe, C.; Maurer, J.; Jacobi, F.K.; Pinckers, A.; et al. The complete form of X-linked congenital stationary night blindness is caused by mutations in a gene encoding a leucine-rich repeat protein. Nat. Genet. 2000, 26, 324–327. [Google Scholar] [CrossRef] [PubMed]
  67. Zeitz, C.; van Genderen, M.; Neidhardt, J.; Luhmann, U.F.; Hoeben, F.; Forster, U.; Wycisk, K.; Mátyás, G.; Hoyng, C.B.; Riemslag, F.; et al. Mutations in GRM6 cause autosomal recessive congenital stationary night blindness with a distinctive scotopic 15-Hz flicker electroretinogram. Investig. Ophthalmol. Vis. Sci. 2005, 46, 4328–4335. [Google Scholar] [CrossRef] [Green Version]
  68. Gregg, R.G.; Mukhopadhyay, S.; Candille, S.I.; Ball, S.L.; Pardue, M.T.; McCall, M.A.; Peachey, N.S. Identification of the gene and the mutation responsible for the mouse nob phenotype. Investig. Ophthalmol. Vis. Sci. 2003, 44, 378–384. [Google Scholar] [CrossRef] [Green Version]
  69. Pinto, L.H.; Vitaterna, M.H.; Shimomura, K.; Siepka, S.M.; McDearmon, E.L.; Balannik, V.; Omura, C.; Lumayag, S.; Invergo, B.M.; Glawe, B.; et al. Generation, Identification and Functional Characterization of the nob4 Mutation of Grm6 in the Mouse. Vis. Neurosci. 2007, 24, 111–123. [Google Scholar] [CrossRef] [Green Version]
  70. Masu, M.; Iwakabe, H.; Tagawa, Y.; Miyoshi, T.; Yamashita, M.; Fukuda, Y.; Sasaki, H.; Hiroi, K.; Nakamura, Y.; Shigemoto, R.; et al. Specific deficit of the ON response in visual transmission by targeted disruption of the mGluR6 gene. Cell 1995, 80, 757–765. [Google Scholar] [CrossRef] [Green Version]
  71. Morgans, C.W.; Brown, R.L.; Duvoisin, R.M. TRPM1: The endpoint of the mGluR6 signal transduction cascade in retinal ON-bipolar cells. BioEssays News Rev. Mol. Cell. Dev. Biol. 2010, 32, 609–614. [Google Scholar] [CrossRef] [PubMed]
  72. Morgans, C.W.; Zhang, J.; Jeffrey, B.G.; Nelson, S.M.; Burke, N.S.; Duvoisin, R.M.; Brown, R.L. TRPM1 is required for the depolarizing light response in retinal ON-bipolar cells. Proc. Natl. Acad. Sci. USA 2009, 106, 19174–19178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Nakamura, M.; Sanuki, R.; Tetsuhiro, R.Y.; Onishi, A.; Nishiguchi, K.M.; Koike, C.; Kadowaki, M.; Kondo, M.; Miyake, Y.; Furukawa, T. TRPM1 mutations are associated with the complete form of congenital stationary night blindness. Mol. Vis. 2010, 16, 425–437. [Google Scholar] [PubMed]
  74. Orhan, E.; Neuillé, M.; de Sousa Dias, M.; Pugliese, T.; Michiels, C.; Condroyer, C.; Antonio, A.; Sahel, J.A.; Audo, I.; Zeitz, C. A New Mouse Model for Complete Congenital Stationary Night Blindness Due to Gpr179 Deficiency. Int. J. Mol. Sci. 2021, 22, 4424. [Google Scholar] [CrossRef] [PubMed]
  75. Orhan, E.; Prézeau, L.; El Shamieh, S.; Bujakowska, K.M.; Michiels, C.; Zagar, Y.; Vol, C.; Bhattacharya, S.S.; Sahel, J.A.; Sennlaub, F.; et al. Further insights into GPR179: Expression, localization, and associated pathogenic mechanisms leading to complete congenital stationary night blindness. Investig. Ophthalmol. Vis. Sci. 2013, 54, 8041–8050. [Google Scholar] [CrossRef] [Green Version]
  76. Peachey, N.S.; Pearring, J.N.; Bojang, P., Jr.; Hirschtritt, M.E.; Sturgill-Short, G.; Ray, T.A.; Furukawa, T.; Koike, C.; Goldberg, A.F.; Shen, Y.; et al. Depolarizing bipolar cell dysfunction due to a Trpm1 point mutation. J. Neurophysiol. 2012, 108, 2442–2451. [Google Scholar] [CrossRef] [Green Version]
  77. Neuillé, M.; Cao, Y.; Caplette, R.; Guerrero-Given, D.; Thomas, C.; Kamasawa, N.; Sahel, J.A.; Hamel, C.P.; Audo, I.; Picaud, S.; et al. LRIT3 Differentially Affects Connectivity and Synaptic Transmission of Cones to ON- and OFF-Bipolar Cells. Investig. Ophthalmol. Vis. Sci. 2017, 58, 1768–1778. [Google Scholar] [CrossRef] [Green Version]
  78. Neuillé, M.; El Shamieh, S.; Orhan, E.; Michiels, C.; Antonio, A.; Lancelot, M.-E.; Condroyer, C.; Bujakowska, K.; Poch, O.; Sahel, J.-A.; et al. Lrit3 Deficient Mouse (nob6): A Novel Model of Complete Congenital Stationary Night Blindness (cCSNB). PLoS ONE 2014, 9, e90342. [Google Scholar] [CrossRef] [Green Version]
  79. Neuillé, M.; Morgans, C.W.; Cao, Y.; Orhan, E.; Michiels, C.; Sahel, J.A.; Audo, I.; Duvoisin, R.M.; Martemyanov, K.A.; Zeitz, C. LRIT3 is essential to localize TRPM1 to the dendritic tips of depolarizing bipolar cells and may play a role in cone synapse formation. Eur. J. Neurosci. 2015, 42, 1966–1975. [Google Scholar] [CrossRef] [Green Version]
  80. Hasan, N.; Pangeni, G.; Ray, T.A.; Fransen, K.M.; Noel, J.; Borghuis, B.G.; McCall, M.A.; Gregg, R.G. LRIT3 is Required for Nyctalopin Expression and Normal ON and OFF Pathway Signaling in the Retina. eNeuro 2020, 7. [Google Scholar] [CrossRef] [Green Version]
  81. Kinoshita, J.; Hasan, N.; Bell, B.A.; Peachey, N.S. Reduced expression of the nob8 gene does not normalize the distribution or function of mGluR6 in the mouse retina. Mol. Vis. 2019, 25, 890–901. [Google Scholar] [PubMed]
  82. Orlandi, C.; Omori, Y.; Wang, Y.; Cao, Y.; Ueno, A.; Roux, M.J.; Condomitti, G.; de Wit, J.; Kanagawa, M.; Furukawa, T.; et al. Transsynaptic Binding of Orphan Receptor GPR179 to Dystroglycan-Pikachurin Complex Is Essential for the Synaptic Organization of Photoreceptors. Cell Rep. 2018, 25, 130–145.e5. [Google Scholar] [CrossRef] [Green Version]
  83. Koike, C.; Obara, T.; Uriu, Y.; Numata, T.; Sanuki, R.; Miyata, K.; Koyasu, T.; Ueno, S.; Funabiki, K.; Tani, A.; et al. TRPM1 is a component of the retinal ON bipolar cell transduction channel in the mGluR6 cascade. Proc. Natl. Acad. Sci. USA 2010, 107, 332–337. [Google Scholar] [CrossRef]
  84. Maddox, D.M.; Vessey, K.A.; Yarbrough, G.L.; Invergo, B.M.; Cantrell, D.R.; Inayat, S.; Balannik, V.; Hicks, W.L.; Hawes, N.L.; Byers, S.; et al. Allelic variance between GRM6 mutants, Grm6nob3 and Grm6nob4 results in differences in retinal ganglion cell visual responses. J. Physiol. 2008, 586, 4409–4424. [Google Scholar] [CrossRef]
  85. Qian, H.; Ji, R.; Gregg, R.G.; Peachey, N.S. Identification of a new mutant allele, Grm6(nob7), for complete congenital stationary night blindness. Vis. Neurosci. 2015, 32, E004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Pardue, M.T.; McCall, M.A.; LaVail, M.M.; Gregg, R.G.; Peachey, N.S. A naturally occurring mouse model of X-linked congenital stationary night blindness. Investig. Ophthalmol. Vis. Sci. 1998, 39, 2443–2449. [Google Scholar]
  87. Chakraborty, R.; Yang, V.; Park, H.N.; Landis, E.G.; Dhakal, S.; Bergen, M.A.; Iuvone, P.M.; Pardue, M.T. Lack of cone mediated retinal function increases susceptibility to form-deprivation myopia in mice. Exp. Eye Res. 2019, 180, 226–230. [Google Scholar] [CrossRef] [Green Version]
  88. Fain, G.; Sampath, A.P. Rod and cone interactions in the retina. F1000Research 2018, 7, F1000. [Google Scholar] [CrossRef]
  89. Tsukamoto, Y.; Omi, N. Classification of Mouse Retinal Bipolar Cells: Type-Specific Connectivity with Special Reference to Rod-Driven AII Amacrine Pathways. Front. Neuroanat. 2017, 11, 92. [Google Scholar] [CrossRef] [PubMed]
  90. Roy, S.; Field, G.D. Dopaminergic modulation of retinal processing from starlight to sunlight. J. Pharmacol. Sci. 2019, 140, 86–93. [Google Scholar] [CrossRef]
  91. Iuvone, P.M.; Tigges, M.; Fernandes, A.; Tigges, J. Dopamine synthesis and metabolism in rhesus monkey retina: Development, aging, and the effects of monocular visual deprivation. Vis. Neurosci. 1989, 2, 465–471. [Google Scholar] [CrossRef]
  92. Stone, R.A.; Lin, T.; Laties, A.M. Muscarinic antagonist effects on experimental chick myopia. Exp. Eye Res. 1991, 52, 755–758. [Google Scholar] [CrossRef]
  93. Feldkaemper, M.P.; Schaeffel, F. Evidence for a potential role of glucagon during eye growth regulation in chicks. Vis. Neurosci. 2002, 19, 755–766. [Google Scholar] [CrossRef]
  94. Troilo, D.; Nickla, D.L.; Mertz, J.R.; Summers Rada, J.A. Change in the synthesis rates of ocular retinoic acid and scleral glycosaminoglycan during experimentally altered eye growth in marmosets. Investig. Ophthalmol. Vis. Sci. 2006, 47, 1768–1777. [Google Scholar] [CrossRef] [PubMed]
  95. Fujikado, T.; Kawasaki, Y.; Fujii, J.; Taniguchi, N.; Okada, M.; Suzuki, A.; Ohmi, G.; Tano, Y. The effect of nitric oxide synthase inhibitor on form-deprivation myopia. Curr. Eye Res. 1997, 16, 992–996. [Google Scholar] [CrossRef] [PubMed]
  96. Mertz, J.R.; Wallman, J. Choroidal retinoic acid synthesis: A possible mediator between refractive error and compensatory eye growth. Exp. Eye Res. 2000, 70, 519–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Landis, E.G.; Chrenek, M.A.; Chakraborty, R.; Strickland, R.; Bergen, M.; Yang, V.; Iuvone, P.M.; Pardue, M.T. Increased endogenous dopamine prevents myopia in mice. Exp. Eye Res. 2020, 193, 107956. [Google Scholar] [CrossRef] [PubMed]
  98. Boatright, J.H.; Gordon, J.R.; Iuvone, P.M. Inhibition of endogenous dopamine release in amphibian retina by L-2-amino-4-phosphonobutyric acid (L-AP4) and trans-2-aminocyclopentane-1,3-dicarboxylate (ACPD). Brain Res. 1994, 649, 339–342. [Google Scholar] [CrossRef] [PubMed]
  99. Aung, M.H.; Hogan, K.; Mazade, R.E.; Park, H.N.; Sidhu, C.S.; Iuvone, P.M.; Pardue, M.T. ON than OFF pathway disruption leads to greater deficits in visual function and retinal dopamine signaling. Exp. Eye Res. 2022, 220, 109091. [Google Scholar] [CrossRef]
  100. Park, H.; Jabbar, S.B.; Tan, C.C.; Sidhu, C.S.; Abey, J.; Aseem, F.; Schmid, G.; Iuvone, P.M.; Pardue, M.T. Visually-driven ocular growth in mice requires functional rod photoreceptors. Investig. Ophthalmol. Vis. Sci. 2014, 55, 6272–6279. [Google Scholar] [CrossRef]
  101. Leifert, D.; Todorova, M.G.; Prünte, C.; Palmowski-Wolfe, A.M. LED-generated multifocal ERG on- and off-responses in complete congenital stationary night blindness—A case report. Doc. Ophthalmol. Adv. Ophthalmol. 2005, 111, 1–6. [Google Scholar] [CrossRef]
  102. Takeuchi, H.; Horie, S.; Moritoh, S.; Matsushima, H.; Hori, T.; Kimori, Y.; Kitano, K.; Tsubo, Y.; Tachibana, M.; Koike, C. Different Activity Patterns in Retinal Ganglion Cells of TRPM1 and mGluR6 Knockout Mice. BioMed Res. Int. 2018, 2018, 2963232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ohtoshi, A.; Wang, S.W.; Maeda, H.; Saszik, S.M.; Frishman, L.J.; Klein, W.H.; Behringer, R.R. Regulation of retinal cone bipolar cell differentiation and photopic vision by the CVC homeobox gene Vsx1. Curr. Biol. 2004, 14, 530–536. [Google Scholar] [CrossRef] [Green Version]
  104. Chakraborty, R.; Park, H.; Aung, M.H.; Tan, C.C.; Sidhu, C.S.; Iuvone, P.M.; Pardue, M.T. Comparison of refractive development and retinal dopamine in OFF pathway mutant and C57BL/6J wild-type mice. Mol. Vis. 2014, 20, 1318–1327. [Google Scholar] [PubMed]
  105. Chakraborty, R.; Pardue, M.T. Molecular and Biochemical Aspects of the Retina on Refraction. Prog. Mol. Biol. Transl. Sci. 2015, 134, 249–267. [Google Scholar]
  106. Tkatchenko, T.V.; Shen, Y.; Tkatchenko, A.V. Analysis of postnatal eye development in the mouse with high-resolution small animal magnetic resonance imaging. Investig. Ophthalmol. Vis. Sci. 2010, 51, 21–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Yang, J.; Yang, L.; Chen, R.; Zhu, Y.; Wang, S.; Hou, X.; Wei, B.; Wang, Q.; Liu, Y.; Qu, J.; et al. A role of color vision in emmetropization in C57BL/6J mice. Sci. Rep. 2020, 10, 14895. [Google Scholar] [CrossRef]
  108. Tkatchenko, T.V.; Shah, R.L.; Nagasaki, T.; Tkatchenko, A.V. Analysis of genetic networks regulating refractive eye development in collaborative cross progenitor strain mice reveals new genes and pathways underlying human myopia. BMC Med. Genom. 2019, 12, 113. [Google Scholar] [CrossRef] [Green Version]
  109. Chakraborty, R.; Ostrin, L.A.; Nickla, D.L.; Iuvone, P.M.; Pardue, M.T.; Stone, R.A. Circadian rhythms, refractive development, and myopia. Ophthalmic Physiol. Opt. J. Br. Coll. Ophthalmic Opt. 2018, 38, 217–245. [Google Scholar] [CrossRef]
  110. Tkatchenko, T.V.; Tkatchenko, A.V. Ketamine-xylazine anesthesia causes hyperopic refractive shift in mice. J. Neurosci. Methods 2010, 193, 67–71. [Google Scholar] [CrossRef] [Green Version]
  111. Glickstein, M.; Millodot, M. Retinoscopy and eye size. Science 1970, 168, 605–606. [Google Scholar] [CrossRef] [PubMed]
  112. Tkatchenko, T.V.; Shen, Y.; Tkatchenko, A.V. Mouse Experimental Myopia Has Features of Primate Myopia. Investig. Ophthalmol. Vis. Sci. 2010, 51, 1297–1303. [Google Scholar] [CrossRef] [Green Version]
  113. Ward, A.H.; Siegwart, J.T.; Frost, M.R.; Norton, T.T. Intravitreally-administered dopamine D2-like (and D4), but not D1-like, receptor agonists reduce form-deprivation myopia in tree shrews. Vis. Neurosci. 2017, 34, E003. [Google Scholar] [CrossRef] [Green Version]
  114. Zhang, S.; Yang, J.; Reinach, P.S.; Wang, F.; Zhang, L.; Fan, M.; Ying, H.; Pan, M.; Qu, J.; Zhou, X. Dopamine Receptor Subtypes Mediate Opposing Effects on Form Deprivation Myopia in Pigmented Guinea Pigs. Investig. Ophthalmol. Vis. Sci. 2018, 59, 4441–4448. [Google Scholar] [CrossRef] [Green Version]
  115. Dong, F.; Zhi, Z.; Pan, M.; Xie, R.; Qin, X.; Lu, R.; Mao, X.; Chen, J.F.; Willcox, M.D.; Qu, J.; et al. Inhibition of experimental myopia by a dopamine agonist: Different effectiveness between form deprivation and hyperopic defocus in guinea pigs. Mol. Vis. 2011, 17, 2824–2834. [Google Scholar] [PubMed]
  116. Feldkaemper, M.; Diether, S.; Kleine, G.; Schaeffel, F. Interactions of spatial and luminance information in the retina of chickens during myopia development. Exp. Eye Res. 1999, 68, 105–115. [Google Scholar] [CrossRef]
  117. Wu, X.H.; Li, Y.Y.; Zhang, P.P.; Qian, K.W.; Ding, J.H.; Hu, G.; Weng, S.J.; Yang, X.L.; Zhong, Y.M. Unaltered retinal dopamine levels in a C57BL/6 mouse model of form-deprivation myopia. Investig. Ophthalmol. Vis. Sci. 2015, 56, 967–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Nickla, D.L. Ocular diurnal rhythms and eye growth regulation: Where we are 50 years after Lauber. Exp. Eye Res. 2013, 114, 25–34. [Google Scholar] [CrossRef] [Green Version]
  119. Qian, K.W.; Li, Y.Y.; Wu, X.H.; Gong, X.; Liu, A.L.; Chen, W.H.; Yang, Z.; Cui, L.J.; Liu, Y.F.; Ma, Y.Y.; et al. Altered Retinal Dopamine Levels in a Melatonin-proficient Mouse Model of Form-deprivation Myopia. Neurosci. Bull. 2022, 38, 992–1006. [Google Scholar] [CrossRef]
  120. Fujikado, T.; Hosohata, J.; Omoto, T. ERG of form deprivation myopia and drug induced ametropia in chicks. Curr. Eye Res. 1996, 15, 79–86. [Google Scholar] [CrossRef] [PubMed]
  121. Crewther, D.P.; Crewther, S.G.; Xie, R.Z. Changes in eye growth produced by drugs which affect retinal ON or OFF responses to light. J. Ocul. Pharmacol. Ther. Off. J. Assoc. Ocul. Pharmacol. Ther. 1996, 12, 193–208. [Google Scholar] [CrossRef] [PubMed]
  122. Gregg, R.G.; Kamermans, M.; Klooster, J.; Lukasiewicz, P.D.; Peachey, N.S.; Vessey, K.A.; McCall, M.A. Nyctalopin expression in retinal bipolar cells restores visual function in a mouse model of complete X-linked congenital stationary night blindness. J. Neurophysiol. 2007, 98, 3023–3033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Gamache, P.H.; Kingery, M.L.; Acworth, I.N. Urinary metanephrine and normetanephrine determined without extraction by using liquid chromatography and coulometric array detection. Clin. Chem. 1993, 39, 1825–1830. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Retinal levels of DA and DOPAC. Quantification of DA (A), DOPAC (B) and the DOPAC/DA ratio (C) in light-adapted adult Gpr179+/+ and Gpr179−/− retinas. Data are expressed as Mean ± SEM. ns: not significant, **: p ≤ 0.01, ***: p < 0.001.
Figure 1. Retinal levels of DA and DOPAC. Quantification of DA (A), DOPAC (B) and the DOPAC/DA ratio (C) in light-adapted adult Gpr179+/+ and Gpr179−/− retinas. Data are expressed as Mean ± SEM. ns: not significant, **: p ≤ 0.01, ***: p < 0.001.
Ijms 24 00219 g001
Figure 2. Natural refractive development. Measurement of the refractive state of Gpr179+/+ and Gpr179−/− mice across age. Data are expressed as Mean ± SEM.
Figure 2. Natural refractive development. Measurement of the refractive state of Gpr179+/+ and Gpr179−/− mice across age. Data are expressed as Mean ± SEM.
Ijms 24 00219 g002
Figure 3. Lens-induction of myopia. Measurement of the kinetics of mean interocular shift between the goggled and ungoggled eyes of Gpr179+/+ and Gpr179−/− mice. Data are expressed as Mean ± SEM. **: p ≤ 0.01, ***: p < 0.001.
Figure 3. Lens-induction of myopia. Measurement of the kinetics of mean interocular shift between the goggled and ungoggled eyes of Gpr179+/+ and Gpr179−/− mice. Data are expressed as Mean ± SEM. **: p ≤ 0.01, ***: p < 0.001.
Ijms 24 00219 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wilmet, B.; Callebert, J.; Duvoisin, R.; Goulet, R.; Tourain, C.; Michiels, C.; Frederiksen, H.; Schaeffel, F.; Marre, O.; Sahel, J.A.; et al. Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia. Int. J. Mol. Sci. 2023, 24, 219. https://doi.org/10.3390/ijms24010219

AMA Style

Wilmet B, Callebert J, Duvoisin R, Goulet R, Tourain C, Michiels C, Frederiksen H, Schaeffel F, Marre O, Sahel JA, et al. Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia. International Journal of Molecular Sciences. 2023; 24(1):219. https://doi.org/10.3390/ijms24010219

Chicago/Turabian Style

Wilmet, Baptiste, Jacques Callebert, Robert Duvoisin, Ruben Goulet, Christophe Tourain, Christelle Michiels, Helen Frederiksen, Frank Schaeffel, Olivier Marre, José Alain Sahel, and et al. 2023. "Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia" International Journal of Molecular Sciences 24, no. 1: 219. https://doi.org/10.3390/ijms24010219

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

Article Metrics

Back to TopTop