Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Neurosci ; 43(48): 8231-8242, 2023 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-37751999

RESUMO

Dopamine is a key neurotransmitter in the signaling cascade controlling ocular refractive development, but the exact role and site of action of dopamine D1 receptors (D1Rs) involved in myopia remains unclear. Here, we determine whether retinal D1Rs exclusively mediate the effects of endogenous dopamine and systemically delivered D1R agonist or antagonist in the mouse form deprivation myopia (FDM) model. Male C57BL/6 mice subjected to unilateral FDM or unobstructed vision were divided into the following four groups: one noninjected and three groups that received intraperitoneal injections of a vehicle, D1R agonist SKF38393 (18 and 59 nmol/g), or D1R antagonist SCH39166 (0.1 and 1 nmol/g). The effects of these drugs on FDM were further assessed in Drd1-knock-out (Drd1-KO), retina-specific conditional Drd1-KO (Drd1-CKO) mice, and corresponding wild-type littermates. In the visually unobstructed group, neither SKF38393 nor SCH39166 affected normal refractive development, whereas myopia development was attenuated by SKF38393 and enhanced by SCH39166 injections. In Drd1-KO or Drd1-CKO mice, however, these drugs had no effect on FDM development, suggesting that activation of retinal D1Rs is pertinent to myopia suppression by the D1R agonist. Interestingly, the development of myopia was unchanged by either Drd1-KO or Drd1-CKO, and neither SKF38393 nor SCH39166 injections, nor Drd1-KO, affected the retinal or vitreal dopamine and the dopamine metabolite DOPAC levels. Effects on axial length were less marked than effects on refraction. Therefore, activation of D1Rs, specifically retinal D1Rs, inhibits myopia development in mice. These results also suggest that multiple dopamine D1R mechanisms play roles in emmetropization and myopia development.SIGNIFICANCE STATEMENT While dopamine is recognized as a "stop" signal that inhibits myopia development (myopization), the location of the dopamine D1 receptors (D1Rs) that mediate this action remains to be addressed. Answers to this key question are critical for understanding how dopaminergic systems regulate ocular growth and refraction. We report here the results of our study showing that D1Rs are essential for controlling ocular growth and myopia development in mice, and for identifying the retina as the site of action for dopaminergic control via D1Rs. These findings highlight the importance of intrinsic retinal dopaminergic mechanisms for the regulation of ocular growth and suggest specific avenues for exploring the retinal mechanisms involved in the dopaminergic control of emmetropization and myopization.


Assuntos
Dopamina , Miopia , Masculino , Camundongos , Animais , Dopamina/metabolismo , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/farmacologia , Camundongos Endogâmicos C57BL , Miopia/genética , Miopia/metabolismo , Retina/metabolismo , Receptores de Dopamina D1/metabolismo
2.
Invest Ophthalmol Vis Sci ; 63(1): 24, 2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-35050306

RESUMO

Purpose: To learn more about the locations of dopamine D2 receptors (D2Rs) that regulate form-deprivation myopia (FDM), using different transgenic mouse models. Methods: One eye of D2R-knockout (KO) mice and wild-type littermates was subjected to four weeks of monocular FDM, whereas the fellow eye served as control. Mice in both groups received daily intraperitoneal injections of either the D2R antagonist sulpiride (8 µg/g) or vehicle alone. FDM was also induced in retina- (Six3creD2Rfl/fl) or fibroblast-specific (S100a4creD2Rfl/fl) D2R-KO mice. A subset of retina-specific D2R-KO mice and D2Rfl/fl littermates were also given sulpiride or vehicle injections. Refraction was measured with an eccentric infrared photorefractor, and other biometric parameters were measured by optical coherence tomography (n ≈ 20 for each group). Results: FDM development was attenuated in wild-type littermates treated with sulpiride. However, this inhibitory effect disappeared in the D2R-KO mice, suggesting that antagonizing D2Rs suppressed myopia development. Similarly, the development of myopia was partially inhibited by retina-specific (deletion efficiency: 94.7%) but not fibroblast-specific (66.9%) D2R-KO. The sulpiride-mediated inhibitory effects on FDM also disappeared with retinal D2R-KO, suggesting that antagonizing D2Rs outside the retina may not attenuate myopia. Changes in axial length were less marked than changes in refraction, but in general the two were correlated. Conclusions: This study demonstrates that D2Rs located in the retina participate in dopaminergic regulation of FDM in mice. These findings provide an important and fundamental basis for further exploring the retinal mechanism(s) involved in dopamine signaling and myopia development.


Assuntos
Regulação da Expressão Gênica , Miopia/genética , RNA/genética , Receptores de Dopamina D2/genética , Retina/metabolismo , Animais , Modelos Animais de Doenças , Eletrorretinografia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Miopia/metabolismo , Miopia/patologia , Receptores de Dopamina D2/biossíntese , Retina/patologia
3.
Neurosci Bull ; 38(9): 992-1006, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35349094

RESUMO

Reduced levels of retinal dopamine, a key regulator of eye development, are associated with experimental myopia in various species, but are not seen in the myopic eyes of C57BL/6 mice, which are deficient in melatonin, a neurohormone having extensive interactions with dopamine. Here, we examined the relationship between form-deprivation myopia (FDM) and retinal dopamine levels in melatonin-proficient CBA/CaJ mice. We found that these mice exhibited a myopic refractive shift in form-deprived eyes, which was accompanied by altered retinal dopamine levels. When melatonin receptors were pharmacologically blocked, FDM could still be induced, but its magnitude was reduced, and retinal dopamine levels were no longer altered in FDM animals, indicating that melatonin-related changes in retinal dopamine levels contribute to FDM. Thus, FDM is mediated by both dopamine level-independent and melatonin-related dopamine level-dependent mechanisms in CBA/CaJ mice. The previously reported unaltered retinal dopamine levels in myopic C57BL/6 mice may be attributed to melatonin deficiency.


Assuntos
Melatonina , Miopia , Animais , Modelos Animais de Doenças , Dopamina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Retina , Privação Sensorial
4.
Invest Ophthalmol Vis Sci ; 62(9): 21, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34259818

RESUMO

Purpose: The purpose of this study was to evaluate the role of the canonical Wnt signaling in the development of the myopia. Methods: Plasma from adult patients with myopia, myopic animal models including the adenomatous polyposis coli (APC) gene mutation mouse model, and the form deprivation (FD) induced mouse model of myopia were used. Niclosamide, a canonical Wnt pathway inhibitor, was orally administrated in animal models. Plasma levels of DKK-1 were determined by using enzyme-linked immunosorbent assay. Refraction, vitreous chamber depth (VCD), axial length (AL), and other parameters, were measured at the end of the FD treatment. Canonical Wnt signaling changes were evaluated by Western blot analysis and immunostaining analysis. Results: Plasma level of Wnt inhibitor DKK-1 was markedly decreased in patients with myopia. Meanwhile, the canonical Wnt pathway was progressively activated during myopia development in mice. Moreover, inhibition of canonical Wnt signaling by niclosamide in mouse models markedly reduced lens thickness (LT), VCD, and AL elongation, resulting in myopia inhibition. Conclusions: Dysregulation of canonical Wnt signaling is a characteristic of myopia and targeting Wnt signaling pathways has potential as a therapeutic strategy for myopia.


Assuntos
Segmento Anterior do Olho/metabolismo , Miopia/genética , Segmento Posterior do Olho/metabolismo , Refração Ocular/fisiologia , Via de Sinalização Wnt/genética , Adolescente , Adulto , Animais , Segmento Anterior do Olho/diagnóstico por imagem , Segmento Anterior do Olho/efeitos dos fármacos , Biomarcadores/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miopia/metabolismo , Miopia/fisiopatologia , Segmento Posterior do Olho/diagnóstico por imagem , Segmento Posterior do Olho/efeitos dos fármacos , Privação Sensorial , Adulto Jovem
5.
Invest Ophthalmol Vis Sci ; 61(6): 47, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32572456

RESUMO

Purpose: The purpose of this study was to explore the role and mechanism of D2 receptor (D2R) involvement in myopia development and the effects of the full D2R agonist quinpirole and partial D2R agonist aripiprazole on postnatal refractive development and form-deprivation myopia (FDM). Methods: C57BL/6 ("B6") mice, raised either in a visually normal or unilateral form-deprivation environment, were divided into three subgroups, including an intraperitoneally injected (IP) vehicle group and two quinpirole (1 and 10 µg/g body weight) treatment groups. The effects of quinpirole on FDM were further verified in D2R-knockout (KO) mice and corresponding wild-type littermates. Then, the modulation of normal vision development and FDM by aripiprazole (1 and 10 µg/g body weight, IP) was assessed in C57BL/6 mice. All biometric parameters were measured before and after treatments, and retinal cyclic adenosine phosphate (cAMP) and phosphorylated ERK (pERK) levels were analyzed to assess D2R-mediated signal transduction. Results: Neither quinpirole nor aripiprazole affected normal refractive development. FDM development was inhibited by quinpirole at low dose but enhanced at high dose, and these bidirectional effects were validated by D2R-specificity. FDM development was attenuated by the partial D2R agonist aripiprazole, at high dose but not at low dose. Quinpirole caused a dose-dependent reduction in cAMP levels, but had no effect on pERK. Aripiprazole reduced cAMP levels at both doses, but caused a dose-dependent increase of pERK in the form-deprived eyes. Conclusions: Reduction of D2R-mediated signaling contributes to myopia development, which can be selectively attenuated by partial D2R agonists that activate D2Rs under the low dopamine levels that occur with FDM.


Assuntos
Miopia/tratamento farmacológico , Miopia/metabolismo , Quimpirol/farmacologia , Receptores de Dopamina D2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Agonistas de Dopamina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Dopamina D2/agonistas
6.
Sci Rep ; 10(1): 14895, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32913294

RESUMO

Spectral composition affects emmetropization in both humans and animal models. Because color vision interacts the effects of chromatic defocus, we developed a method to bypass the effects of longitudinal chromatic aberration by placing a spectral filter behind the optics of the eye, using genetic tools. Newborn C57BL/6J (B6) mice were reared in quasi-monochromatic red (410-510 nm) or blue (585-660 nm) light beginning before eye-opening. Refractive states and ocular dimensions were compared at 4, 6, 8, and 10 weeks with mice reared in normal white light. Cre recombinase-dependent Ai9 reporter mice were crossed with Chx10-Cre to obtain Chx10-Cre;Ai9 mice, expressing red fluorescent protein in retinal Cre-positive cells. Ai9 offsprings, with and without Cre, were reared under a normal visual environment. Refraction and axial components were measured as described above. Expression levels of M and S opsin were quantified by western blotting at 10 weeks. Compared with those reared in white light, B6 mice reared in red light developed relative hyperopia, principally characterized by flattening of corneal curvature. Emmetropization was not affected by blue light, possibly because the reduction in vitreous chamber depth compensated for the increase in corneal curvature. Compared with Cre-negative littermates, the refraction and axial dimensions of Chx10-Cre;Ai9 mice were not significantly different at the follow-up timepoints. M opsin levels were higher in Chx10-Cre;Ai9 mice at 10 weeks while S opsin levels were not different. Red light induced a hyperopic shift in mouse refractive development. Emmetropization was not impacted in mice with perturbed color vision caused by intrinsic red-fluorescent protein, suggesting that color vision may not be necessary in mouse emmetropization when other mechanisms are present.


Assuntos
Visão de Cores , Emetropia/fisiologia , Animais , Eletrorretinografia , Camundongos , Camundongos Endogâmicos C57BL , Refração Ocular , Retina/fisiologia
7.
Brain Res ; 1726: 146537, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31672473

RESUMO

Higher visual centers could modulate visually-guided ocular growth, in addition to local mechanisms intrinsic to the eye. There is evidence that such central modulations could be species (even subspecies)-dependent. While the mouse has recently become an important experimental animal in myopia studies, it remains unclear whether and how visual centers modulate refractive development in mice, an issue that was examined in the present study. We found that optic nerve crush (ONC), performed at P18, could modify normal refractive development in the C57BL/6 mouse raised in normal visual environment. Unexpectedly, sham surgery caused a steeper cornea, leading to a modest myopic refractive shift, but did not induce significant changes in ocular axis length. ONC caused corneal flattening and re-calibrated the refractive set-point in a bidirectional manner, causing significant myopic (<-3 D, 54.5%) or hyperopic (>+3 D, 18.2%) shifts in refractive error in most (totally 72.7%) animals, both due to changes in ocular axial length. ONC did not change the density of dopaminergic amacrine cells, but increased retinal levels of dopamine and DOPAC. We conclude that higher visual centers are likely to play a role in fine-tuning of ocular growth, thus modifying refractive development in the C57BL/6 mouse. The changes in refractive error induced by ONC are accounted for by alternations in multiple ocular dimensions, including corneal curvature and axial length.


Assuntos
Miopia/fisiopatologia , Nervo Óptico/crescimento & desenvolvimento , Retina/crescimento & desenvolvimento , Vias Visuais/crescimento & desenvolvimento , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Células Amácrinas/metabolismo , Animais , Córnea/crescimento & desenvolvimento , Córnea/patologia , Dopamina/metabolismo , Camundongos Endogâmicos C57BL , Miopia/metabolismo , Miopia/patologia , Compressão Nervosa , Retina/metabolismo , Retina/patologia , Tirosina 3-Mono-Oxigenase/metabolismo , Vias Visuais/metabolismo
8.
EBioMedicine ; 57: 102878, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32652319

RESUMO

BACKGROUND: Myopia is a good model for understanding the interaction between genetics and environmental stimuli. Here we dissect the biological processes affecting myopia progression. METHODS: Human Genetic Analyses: (1) gene set analysis (GSA) of new genome wide association study (GWAS) data for 593 individuals with high myopia (refraction ≤ -6 diopters [D]); (2) over-representation analysis (ORA) of 196 genes with de novo mutations, identified by whole genome sequencing of 45 high-myopia trio families, and (3) ORA of 284 previously reported myopia risk genes. Contributions of the enriched signaling pathways in mediating the genetic and environmental interactions during myopia development were investigated in vivo and in vitro. RESULTS: All three genetic analyses showed significant enrichment of four KEGG signaling pathways, including amphetamine addiction, extracellular matrix (ECM) receptor interaction, neuroactive ligand-receptor interaction, and regulation of actin cytoskeleton pathways. In individuals with extremely high myopia (refraction ≤ -10 D), the GSA of GWAS data revealed significant enrichment of the HIF-1α signaling pathway. Using human scleral fibroblasts, silencing the key nodal genes within protein-protein interaction networks for the enriched pathways antagonized the hypoxia-induced increase in myofibroblast transdifferentiation. In mice, scleral HIF-1α downregulation led to hyperopia, whereas upregulation resulted in myopia. In human subjects, near work, a risk factor for myopia, significantly decreased choroidal blood perfusion, which might cause scleral hypoxia. INTERPRETATION: Our study implicated the HIF-1α signaling pathway in promoting human myopia through mediating interactions between genetic and environmental factors. FUNDING: National Natural Science Foundation of China grants; Natural Science Foundation of Zhejiang Province.


Assuntos
Interação Gene-Ambiente , Predisposição Genética para Doença , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Miopia/genética , Animais , Modelos Animais de Doenças , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Camundongos , Miopia/metabolismo , Miopia/patologia , Esclera/metabolismo , Esclera/patologia , Transdução de Sinais
9.
Invest Ophthalmol Vis Sci ; 59(6): 2623-2634, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29847669

RESUMO

Purpose: To determine the roles of dopamine D2 receptors (D2Rs) and dopamine D1 receptors (D1Rs) in the inhibition of form-deprivation myopia (FDM) by the nonselective dopamine agonist apomorphine (APO) in D2R-knockout (D2R-KO) and D1R-KO mice. Methods: Retinal layer thicknesses and electroretinograms (ERGs) were analyzed in KO mice and in D2R and D1R antagonist-treated mice. D2R-KO or D1R-KO mice and wild-type (WT) littermates were subjected to form deprivation during postnatal weeks 5 to 8. Both groups were intraperitoneally injected daily with either APO (5 µg/g body weight) dissolved in 1 µg/µL ascorbic acid or vehicle alone. Refraction, vitreous chamber depth (VCD), and axial length (AL), among other parameters, were measured prior to and at the end of the treatment period. Results: The retinal layer thicknesses and ERGs in KO mice were similar to those treated with D2R and D1R antagonists. APO administration in WT mice inhibited the development of FDM by approximately 80%. FDM in D2R-KO mice was inhibited approximately 50% compared with WT mice and was further inhibited by APO to a level similar to that in APO-treated WT mice. FDM development in D1R-KO mice was similar to that in WT mice and was not affected by APO administration. The changes in VCD and AL were consistent with refraction data. Conclusions: In mice, APO-mediated FDM inhibition was abolished by D1R KO but not D2R KO. This indicates the specificity of D1Rs for the pharmacologic inhibitory effect of APO on FDM and a nonessential role of D2Rs in this process in mice.


Assuntos
Apomorfina/farmacologia , Agonistas de Dopamina/farmacologia , Miopia/tratamento farmacológico , Miopia/fisiopatologia , Receptores de Dopamina D1/fisiologia , Retina/fisiopatologia , Animais , Modelos Animais de Doenças , Eletrorretinografia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Retina/efeitos dos fármacos , Privação Sensorial
11.
PLoS One ; 10(10): e0141144, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26495845

RESUMO

Myopia incidence in China is rapidly becoming a very serious sight compromising problem in a large segment of the general population. Therefore, delineating the underlying mechanisms leading to myopia will markedly lessen the likelihood of other sight compromising complications. In this regard, there is some evidence that patients afflicted with familial adenomatous polyposis (FAP), havean adenomatous polyposis coli (APC) mutation and a higher incidence of myopia. To clarify this possible association, we determined whether the changes in pertinent biometric and biochemical parameters underlying postnatal refractive error development in APCMin mice are relevant for gaining insight into the pathogenesis of this disease in humans. The refraction and biometrics in APCMin mice and age-matched wild-type (WT) littermates between postnatal days P28 and P84 were examined with eccentric infrared photorefraction (EIR) and customized optical coherence tomography (OCT). Compared with WT littermates, the APCMin mutated mice developed myopia (average -4.64 D) on P84 which was associated with increased vitreous chamber depth (VCD). Furthermore, retinal and scleral changes appear in these mice along with: 1) axial length shortening; 2) increased retinal cell proliferation; 3) and decreased tyrosine hydroxylase (TH) expression, the rate-limiting enzyme of DA synthesis. Scleral collagen fibril diameters became heterogeneous and irregularly organized in the APCMin mice. Western blot analysis showed that scleral alpha-1 type I collagen (col1α1) expression also decreased whereas MMP2 and MMP9 mRNA expression was invariant. These results indicate that defective APC gene function promotes refractive error development. By characterizing in APCMin mice ocular developmental changes, this approach provides novel insight into underlying pathophysiological mechanisms contributing to human myopia development.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Miopia/genética , Retina/metabolismo , Esclera/metabolismo , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Miopia/metabolismo , Miopia/patologia , Refração Ocular , Retina/patologia , Esclera/patologia , Tomografia de Coerência Óptica , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Visão Ocular , Corpo Vítreo/metabolismo , Corpo Vítreo/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA