Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Neurosci ; 41(12): 2566-2580, 2021 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-33536197

RESUMEN

We show for the first time that the neuropeptide orexin modulates pupillary light response, a non-image-forming visual function, in mice of either sex. Intravitreal injection of the orexin receptor (OXR) antagonist TCS1102 and orexin-A reduced and enhanced pupillary constriction in response to light, respectively. Orexin-A activated OX1Rs on M2-type intrinsically photosensitive retinal ganglion cells (M2 cells), and caused membrane depolarization of these cells by modulating inward rectifier potassium channels and nonselective cation channels, thus resulting in an increase in intrinsic excitability. The increased intrinsic excitability could account for the orexin-A-evoked increase in spontaneous discharges and light-induced spiking rates of M2 cells, leading to an intensification of pupillary constriction. Orexin-A did not alter the light response of M1 cells, which could be because of no or weak expression of OX1Rs on them, as revealed by RNAscope in situ hybridization. In sum, orexin-A is likely to decrease the pupil size of mice by influencing M2 cells, thereby improving visual performance in awake mice via enhancing the focal depth of the eye's refractive system.SIGNIFICANCE STATEMENT This study reveals the role of the neuropeptide orexin in mouse pupillary light response, a non-image-forming visual function. Intravitreal orexin-A administration intensifies light-induced pupillary constriction via increasing the excitability of M2 intrinsically photosensitive retinal ganglion cells by activating the orexin receptor subtype OX1R. Modulation of inward rectifier potassium channels and nonselective cation channels were both involved in the ionic mechanisms underlying such intensification. Orexin could improve visual performance in awake mice by reducing the pupil size and thereby enhancing the focal depth of the eye's refractive system.


Asunto(s)
Orexinas/administración & dosificación , Estimulación Luminosa/métodos , Pupila/efectos de los fármacos , Reflejo Pupilar/efectos de los fármacos , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Bencimidazoles/administración & dosificación , Femenino , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Orexina/agonistas , Receptores de Orexina/metabolismo , Orexinas/antagonistas & inhibidores , Pupila/fisiología , Pirrolidinas/administración & dosificación , Reflejo Pupilar/fisiología , Células Ganglionares de la Retina/metabolismo
2.
Exp Eye Res ; 200: 108240, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32919994

RESUMEN

Aerobic exercise has been shown to play a crucial role in preventing neurological diseases and improving cognitive function. In the present study, we investigated the effect of treadmill training on retinal ganglion cells (RGCs) following optic nerve transection in adult rats. We exercised the rats on a treadmill for 5 d/week (30 min/d at a rate of 9 m/min) or placed control rats on static treadmills. After 3 weeks of exercise, the left optic nerve of each rat was transected. After the surgery, the rat was exercised for another week. The percentages of surviving RGCs in the axotomized eyes of inactive rats were 67% and 39% at 5 and 7 days postaxotomy, respectively. However, exercised rats had significant more RGCs at 5 (74% survival) and 7 days (48% survival) after axotomy. Moreover, retinal brain-derived neurotrophic factor (BDNF) protein levels were significantly upregulated in response to exercise compared with those in the axotomized eyes of inactive rats. Blocking BNDF signaling during exercise by intraperitoneal injections of ANA-12, a BDNF tropomyosin receptor kinase (TrkB) receptor antagonist, reduced the number of RGCs in exercised rats to the level of RGCs in the inactive rats, effectively abolishing the protection of RGCs afforded by exercise. The results suggest that treadmill training effectively rescues RGCs from neurodegeneration following optic nerve transection by upregulating the expression of BDNF.


Asunto(s)
Traumatismos del Nervio Óptico/patología , Nervio Óptico/patología , Condicionamiento Físico Animal/métodos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Muerte Celular , Modelos Animales de Enfermedad , Masculino , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/terapia , Ratas , Regulación hacia Arriba
3.
Exp Eye Res ; 188: 107812, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31550445

RESUMEN

In this work, we investigated changes in the morphology of intrinsically photosensitive retinal ganglion cells (ipRGCs), M1 subtype, and pupillary light reflex following local and selective ablation of photoreceptors in mice. Laser photocoagulation was used to selectively destroy four patches of photoreceptors per eye at around 4 papillary diameters from the optic disc and at the 3, 6, 9, and 12 o'clock positions between the retinal vessels in the adult mouse retina, leaving cells in the inner retina intact. Morphological parameters of individual M1 cells specifically labeled by the antibody against melanopsin (PA1-780), including dendritic field size, total dendritic length, and dendritic branch number, were examined 1, 2, 4, and 8 weeks after photocoagulation with Neurolucida software. A considerable reduction in these parameters in M1 cells in the "lesioned areas" was found at all the four time points after photocoagulation, as compared with those in the "unlesioned areas". Although M1 cells in the lesioned areas showed significant changes as early as 1 week after laser treatment and the changes gradually increased, reaching a peak value at 2 weeks, morphological restoration was clearly seen in these cells over time. However, no difference in the morphological parameters of M1 cells was observed between the unlesioned areas of laser-treated mice and the corresponding areas of age-matched normal mice without laser lesions. Fluorescence intensity of the somata of melanopsin-positive M1 cells located inside the lesioned areas was significantly decreased at all the four time points after photocoagulation, whereas no changes in pupillary light reflex were detected at different light irradiations, indicating that photocoagulation-induced local photoreceptor loss and alterations of ipRGCs may be insufficient to cause abnormalities in non-image-forming (NIF) visual functions. The results suggest that intact photoreceptors could be crucial for maintaining the expression levels of melanopsin and normal morphology of M1 cells.


Asunto(s)
Coagulación con Láser , Reflejo Pupilar/fisiología , Retina/cirugía , Células Ganglionares de la Retina/patología , Animales , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras de Vertebrados/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/efectos de la radiación , Opsinas de Bastones/metabolismo
4.
Sheng Li Xue Bao ; 68(2): 135-40, 2016 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-27108899

RESUMEN

Due to the advantages in genetic manipulation, mice have become one of the most commonly used mammalian models for the study of mechanisms underlying myopia development. However, the vast majority of laboratory mouse strains are incapable of synthesizing melatonin, a neurohormone that may play an important role in myopia generation in humans. The present study investigated refractive development profiles in the CBA/CaJ mouse, a strain proficient in melatonin, and determined whether and how its refractive development could be affected by form-deprivation. Eccentric infrared photoretinoscopy revealed that this animal could be stably refracted, and the refractive error underwent developmental changes, which increased with age in the hyperopic direction and eventually got stable approximately 9 weeks after birth. The absolute values of refractive error in CBA/CaJ mice were larger than those of age-matched C57BL/6 mice, whereas the time points when refractive error reached steady state were similar between the two strains. Five weeks of form-deprivation applied to 3-week-old CBA/CaJ mice by translucent occluder wear caused a significant myopic shift in refractive error, indicating that this strain could be adequately used as a myopia model.


Asunto(s)
Refracción Ocular , Privación Sensorial , Animales , Modelos Animales de Enfermedad , Ojo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Miopía
5.
Neural Regen Res ; 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38934389

RESUMEN

ABSTRACT: Diabetic retinopathy is a prominent cause of blindness in adults, with early retinal ganglion cell (RGC) loss contributing to visual dysfunction or blindness. In the brain, defects in y-aminobutyric acid (GABA) synaptic transmission are associated with pathophysiological and neurodegenerative disorders, whereas glucagon-like peptide-1 (GLP-1) has demonstrated neuroprotective effects. However, it is not yet clear whether diabetes causes alterations in inhibitory input to RGCs and whether and how GLP-1 protects against neurodegeneration in the diabetic retina through regulating inhibitory synaptic transmission to RGCs. In the present study, we used the patch-clamp technique to record GABA subtype A receptor-mediated miniature inhibitory postsynaptic currents (mIPSCs) in RGCs from streptozotocin-induced diabetes model rats. We found that early diabetes (4 weeks of hyperglycemia) decreased the frequency of GABAergic mIPSCs in RGCs without altering their amplitude, suggesting a reduction in the spontaneous release of GABA to RGCs. Topical administration of GLP-1 eyedrops over a period of 2 weeks effectively countered the hyperglycemia-induced downregulation of GABAergic mIPSC frequency, subsequently enhancing the survival of RGCs. Concurrently, the protective effects of GLP-1 on RGCs in diabetic rats were eliminated by topical administration of exendin-9-39, a specific GLP-1 receptor antagonist, or SR95531, a specific antagonist of the GABA subtype A receptor. Furthermore, extracellular perfusion of GLP-1 was found to elevate the frequencies of GABAergic mIPSCs in both ON- and OFF-type RGCs. This elevation was shown to be mediated by activation of the phosphatidylinositol-phospholipase C/inositol 1,4,5-trisphosphate receptor/Ca2+/protein kinase C signaling pathway downstream of GLP-1 receptor activation. Moreover, multielectrode array recordings revealed that GLP-1 functionally augmented the photoresponses of ON-type RGCs. Optomotor response tests demonstrated that diabetic rats exhibited reductions in visual acuity and contrast sensitivity that were significantly ameliorated by topical administration of GLP-1. These results suggest that GLP-1 facilitates the release of GABA onto RGCs through the activation of GLP-1 receptor, leading to the de-excitation of RGC circuits and the inhibition of excitotoxic processes associated with diabetic retinopathy. Collectively, our findings indicate that the GABA system has potential as a therapeutic target for mitigating early-stage diabetic retinopathy. Furthermore, the topical administration of GLP-1 eyedrops represents a non-invasive and effective treatment approach for managing early-stage diabetic retinopathy.

6.
iScience ; 26(9): 107680, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37680468

RESUMEN

Progressive damage of retinal ganglion cells (RGCs) is observed in early diabetic retinopathy. Intracellular Ca2+ overload mediated by Ca2+ influx through voltage-gated Ca2+ channels (VGCCs) is involved in neurodegeneration, whereas glucagon-like peptide-1 (GLP-1) provides neuroprotection. However, whether GLP-1 plays a neuroprotective role in diabetic retinas by modulating VGCCs remains unknown. We found that eye drops of exendin-4, a long-acting GLP-1 receptor (GLP-1R) agonist, prevented the increase of L-type Ca2+ current (ILCa) densities of RGCs induced by 4-week hyperglycemia and promoted RGC survival by suppressing L-type VGCC (L-VGCC) activity in streptozotocin-induced diabetic rats. Moreover, exendin-4-induced suppression of ILCa in RGCs may be mediated by a GLP-1R/Gs/cAMP-PKA/ryanodine/Ca2+/calmodulin/calcineurin/PP1 signaling pathway. Furthermore, exendin-4 functionally improved the light-evoked spiking ability of diabetic RGCs. These results suggest that GLP-1R activation enhances cAMP to PP1 signaling and that PP1 inactivates L-VGCCs by dephosphorylating them, thereby reducing Ca2+ influx, which could protect RGCs against excitotoxic Ca2+ overload.

7.
Sci Adv ; 9(12): eadf4651, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36947616

RESUMEN

Light modulates mood through various retina-brain pathways. We showed that mice treated with short-term acute bright light exposure displayed anxiety-related phenotypes in a prolonged manner even after the termination of the exposure. Such a postexposure anxiogenic effect depended upon melanopsin-based intrinsically photosensitive retinal ganglion cell (ipRGC) activities rather than rod/cone photoreceptor inputs. Chemogenetic manipulation of specific central nuclei demonstrated that the ipRGC-central amygdala (CeA) visual circuit played a key role in this effect. The corticosterone system was likely to be involved in this effect, as evidenced by enhanced expression of the glucocorticoid receptor (GR) protein in the CeA and the bed nucleus of the stria terminalis and by the absence of this effect in animals treated with the GR antagonist. Together, our findings reveal a non-image forming visual circuit specifically designed for "the delayed" extinction of anxiety against potential threats, thus conferring a survival advantage.


Asunto(s)
Núcleo Amigdalino Central , Células Ganglionares de la Retina , Ratones , Animales , Células Ganglionares de la Retina/metabolismo , Retina , Células Fotorreceptoras Retinianas Conos , Células Fotorreceptoras de Vertebrados/metabolismo , Luz
8.
Neurosci Bull ; 38(6): 622-636, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35278196

RESUMEN

Glucagon-like peptide-1 (GLP-1) is expressed in retinal neurons, but its role in the retina is largely unknown. Here, we demonstrated that GLP-1 or the GLP-1 receptor (GLP-1R; a G protein-coupled receptor) agonist exendin-4 suppressed γ-aminobutyric acid receptor (GABAR)-mediated currents through GLP-1Rs in isolated rat retinal ganglion cells (GCs). Pre-incubation with the stimulatory G protein (Gs) inhibitor NF 449 abolished the exendin-4 effect. The exendin-4-induced suppression was mimicked by perfusion with 8-Br-cAMP (a cAMP analog), but was eliminated by the protein kinase A (PKA) inhibitor Rp-cAMP/KT-5720. The exendin-4 effect was accompanied by an increase in [Ca2+]i of GCs through the IP3-sensitive pathway and was blocked in Ca2+-free solution. Furthermore, when the activity of calmodulin (CaM) and CaM-dependent protein kinase II (CaMKII) was inhibited, the exendin-4 effect was eliminated. Consistent with this, exendin-4 suppressed GABAR-mediated light-evoked inhibitory postsynaptic currents in GCs in rat retinal slices. These results suggest that exendin-4-induced suppression may be mediated by a distinct Gs/cAMP-PKA/IP3/Ca2+/CaM/CaMKII signaling pathway, following the activation of GLP-1Rs.


Asunto(s)
Péptido 1 Similar al Glucagón , Células Ganglionares de la Retina , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Exenatida/metabolismo , Exenatida/farmacología , Péptido 1 Similar al Glucagón/farmacología , Ratas , Receptores de GABA/metabolismo , Células Ganglionares de la Retina/fisiología , Transducción de Señal
9.
Sci Adv ; 8(23): eabm9027, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35675393

RESUMEN

The increasing global prevalence of myopia calls for elaboration of the pathogenesis of this disease. Here, we show that selective ablation and activation of intrinsically photosensitive retinal ganglion cells (ipRGCs) in developing mice induced myopic and hyperopic refractive shifts by modulating the corneal radius of curvature (CRC) and axial length (AL) in an opposite way. Melanopsin- and rod/cone-driven signals of ipRGCs were found to influence refractive development by affecting the AL and CRC, respectively. The role of ipRGCs in myopia progression is evidenced by attenuated form-deprivation myopia magnitudes in ipRGC-ablated and melanopsin-deficient animals and by enhanced melanopsin expression/photoresponses in form-deprived eyes. Cell subtype-specific ablation showed that M1 subtype cells, and probably M2/M3 subtype cells, are involved in ocular development. Thus, ipRGCs contribute substantially to mouse eye growth and myopia development, which may inspire novel strategies for myopia intervention.


Asunto(s)
Miopía , Células Ganglionares de la Retina , Animales , Ratones , Miopía/etiología , Células Fotorreceptoras de Vertebrados , Células Ganglionares de la Retina/fisiología , Visión Ocular
10.
Neurosci Bull ; 38(9): 992-1006, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35349094

RESUMEN

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.


Asunto(s)
Melatonina , Miopía , Animales , Modelos Animales de Enfermedad , Dopamina , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Retina , Privación Sensorial
11.
Neurosignals ; 19(2): 110-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21555866

RESUMEN

Using patch-clamp whole-cell recording, we investigated how activation of the sigma receptor 1 (σR1) modulates light-evoked excitatory postsynaptic currents (eEPSCs) of ganglion cells (GCs) in rat retinal slice preparations. Bath application of the σR1 agonist SKF10047 (SKF) suppressed N-methyl-D-aspartate (NMDA) receptor-mediated eEPSCs at different holding potentials in ON, OFF and ON-OFF GCs, and the effects were blocked when the preparations were pre-incubated with the σR1 antagonist BD1047. In contrast, SKF had no effects on α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-mediated eEPSCs of these GCs. Furthermore, application of SKF did not affect AMPA receptor-mediated miniature EPSCs of GCs, suggesting that activation of σR1 did not change the release of glutamate from bipolar cells. These results suggest that σR1 may be involved in the regulation of output signaling of GCs by preferentially modulating NMDA receptor-mediated eEPSCs of these retinal neurons.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Luz , Receptores de N-Metil-D-Aspartato/fisiología , Receptores sigma/fisiología , Células Ganglionares de la Retina/fisiología , Transmisión Sináptica/fisiología , Visión Ocular/fisiología , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Receptor Cross-Talk/fisiología , Receptores sigma/agonistas , Receptores sigma/antagonistas & inhibidores , Transducción de Señal/fisiología
12.
Neuropharmacology ; 187: 108492, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33582153

RESUMEN

In this work, modulation by orexin-A of the release of glutamate and GABA from bipolar and amacrine cells respectively was studied by examining the effects of the neuropeptide on miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) of rat retinal ganglion cells (GCs). Using RNAscope in situ hybridization in combination with immunohistochemistry, we showed positive signals for orexin receptor-1 (OX1R) mRNA in the bipolar cell terminals and those for orexin receptor-2 (OX2R) mRNA in the amacrine cell terminals. With whole-cell patch-clamp recordings in rat retinal slices, we demonstrated that application of orexin-A reduced the interevent interval of mEPSCs of GCs through OX1R. However, it increased the interevent interval of mIPSCs, mediated by GABAA receptors, through OX2R. Furthermore, orexin-A-induced reduction of mEPSC interevent interval was abolished by the application of PI-PLC inhibitors or PKC inhibitors. In contrast, orexin-A-induced increase of GABAergic mIPSC interevent interval was mimicked by 8-Br-cAMP or an adenylyl cyclase activator, but was eliminated by PKA antagonists. Finally, application of nimodipine, an L-type Ca2+ channel blocker, increased both mEPSC and mIPSC interevent interval, and co-application of orexin-A no longer changed the mEPSCs and mIPSCs. We conclude that orexin-A increases presynaptic glutamate release onto GCs by activating L-type Ca2+ channels in bipolar cells, a process that is mediated by an OX1R/PI-PLC/PKC signaling pathway. However, orexin-A decreases presynaptic GABA release onto GCs by inhibiting L-type Ca2+ channels in amacrine cells, a process that is mediated by an OX2R/cAMP-PKA signaling pathway.


Asunto(s)
Células Amacrinas/metabolismo , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Inhibidores/genética , Receptores de Orexina/genética , Orexinas/metabolismo , Células Bipolares de la Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Transmisión Sináptica/genética , Células Amacrinas/efectos de los fármacos , Animales , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Glutámico/efectos de los fármacos , Ácido Glutámico/metabolismo , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Receptores de Orexina/metabolismo , Orexinas/farmacología , Técnicas de Placa-Clamp , Fosfoinositido Fosfolipasa C/antagonistas & inhibidores , Fosfoinositido Fosfolipasa C/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Ratas , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Células Bipolares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
13.
Diabetes ; 70(5): 1157-1169, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33574020

RESUMEN

Recent evidence suggests that melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs), a neuronal class regulating nonimage forming (NIF) vision and generally thought to be injury resistant, are dysfunctional in certain neurodegenerative diseases. Although disrupted NIF visual functions have been reported in patients and animals with diabetes, it remains controversial whether ipRGCs exhibit remodeling during diabetes and if so, whether such remodeling is variable among ipRGC subtypes. Here, we demonstrate that survival, soma-dendritic profiles, and melanopsin-based functional activity of M1 ipRGCs were unaltered in streptozotocin-induced 3-month diabetic mice. Such resistance remained at 6 months after streptozotocin administration. In contrast, M2/M3 ipRGCs underwent significant remodeling in diabetic mice, manifested by enlarged somata and increased dendritic branching complexity. Consistent with the unaltered melanopsin levels, the sensitivity of melanopsin-based activity was unchanged in surviving M2 cells, but their response gain displayed a compensatory enhancement. Meanwhile, the pupillary light reflex, a NIF visual function controlled by M2 cells, was found to be impaired in diabetic animals. The resistance of M1 cells might be attributed to the adjacency of their dendrites to capillaries, which makes them less disturbed by the impaired retinal blood supply at the early stage of diabetes.


Asunto(s)
Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Retina/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Opsinas de Bastones/metabolismo , Estreptozocina/toxicidad , Animales , Ratones , Retina/efectos de los fármacos
14.
Cells ; 9(12)2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33271919

RESUMEN

Correlated spontaneous activity in the developing retina (termed "retinal waves") plays an instructive role in refining neural circuits of the visual system. Depolarizing (ON) and hyperpolarizing (OFF) starburst amacrine cells (SACs) initiate and propagate cholinergic retinal waves. Where cholinergic retinal waves stop, SACs are thought to be driven by glutamatergic retinal waves initiated by ON-bipolar cells. However, the properties and function of cholinergic and glutamatergic waves in ON- and OFF-SACs still remain poorly understood. In the present work, we performed whole-cell patch-clamp recordings and Ca2+ imaging from genetically labeled ON- and OFF-SACs in mouse flat-mount retinas. We found that both SAC subtypes exhibited spontaneous rhythmic depolarization during cholinergic and glutamatergic waves. Interestingly, ON-SACs had wave-induced action potentials (APs) in an age-dependent manner, but OFF-SACs did not. Simultaneous Ca2+ imaging and patch-clamp recordings demonstrated that, during a cholinergic wave, APs of an ON-SAC appeared to promote the dendritic release of acetylcholine onto neighboring ON- and OFF-SACs, which enhances their Ca2+ transients. These results advance the understanding of the cellular mechanisms underlying correlated spontaneous activity in the developing retina.


Asunto(s)
Acetilcolina/metabolismo , Potenciales de Acción/fisiología , Células Amacrinas/metabolismo , Células Amacrinas/fisiología , Glutamatos/metabolismo , Retina/fisiología , Animales , Animales Recién Nacidos , Calcio/metabolismo , Colinérgicos/metabolismo , Femenino , Masculino , Ratones , Retina/metabolismo
15.
Invest Ophthalmol Vis Sci ; 61(2): 14, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32049344

RESUMEN

Purpose: Experimental access to specific cell subtypes is essential for deciphering the complexity of retinal networks. Here, we characterized the selective labeling, caused by ectopic transgene expression, of two atypical retinal neurons in the ChAT-Channelrhodopsin-2 (ChR2)-EYFP mouse. Methods: Retinal sections and flat-mounts were prepared for double-staining immunohistochemistry with antibodies against EYFP and various neuronal markers. Sagittal/coronal brain slices were made to visualize EYFP signals in central nuclei. Whole-cell recordings were conducted to test the functionality of ChR2. Results: Two populations of EYFP-positive retinal cells were observed. The inner nuclear layer (INL)-located one (type I cell) distributed regularly throughout the entire retina, whereas the ganglion cell layer (GCL)-residing one (type II cell) was restricted ventrally. None of them was cholinergic, as evidenced by the complete absence of ChAT immunoreactivity. Type I cells were immunolabeled by the amacrine marker syntaxin. However, the vast majority of them were neither positive to GABA/GAD65, nor to GlyT1/glycine, suggesting that they were non-GABAergic non-glycinergic amacrine cells (nGnG ACs), which was confirmed by double-labeling with the nGnG AC marker PPP1R17. Type II cells were immunopositive to melanopsin, but not to Brn3a or Brn3b. They possessed dendrites stratifying in the outermost inner plexiform layer (IPL) and axons projecting to the suprachiasmatic nucleus (SCN) rather than the olivary pretectal nucleus (OPN), suggesting that they belonged to a Brn3b-negative subset of M1-type intrinsically photosensitive retinal ganglion cells (ipRGCs). Glutamatergic transmission-independent photocurrents were elicited in EYFP-positive cells, indicating the functional expression of ChR2. Conclusions: The ChAT-ChR2-EYFP retina exhibits ectopic, but functional, transgene expression in nGnG ACs and SCN-innervating M1 ipRGCs, thus providing an ideal tool to achieve efficient labeling and optogenetic manipulation of these cells.


Asunto(s)
Células Amacrinas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3B/metabolismo , Transgenes/fisiología , Animales , Channelrhodopsins/metabolismo , Colina O-Acetiltransferasa/metabolismo , Femenino , Expresión Génica , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Transgenes/genética
16.
Brain Res ; 1726: 146537, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31672473

RESUMEN

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.


Asunto(s)
Miopía/fisiopatología , Nervio Óptico/crecimiento & desarrollo , Retina/crecimiento & desarrollo , Vías Visuales/crecimiento & desarrollo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Células Amacrinas/metabolismo , Animales , Córnea/crecimiento & desarrollo , Córnea/patología , Dopamina/metabolismo , Ratones Endogámicos C57BL , Miopía/metabolismo , Miopía/patología , Compresión Nerviosa , Retina/metabolismo , Retina/patología , Tirosina 3-Monooxigenasa/metabolismo , Vías Visuales/metabolismo
17.
Invest Ophthalmol Vis Sci ; 60(5): 1644-1656, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30995300

RESUMEN

Purpose: We investigate morphologic and physiologic alterations of ganglion cells (GCs) in a streptozocin (STZ)-induced diabetic mouse model. Methods: Experiments were conducted in flat-mount retinas of mice 3 months after the induction of diabetes. Changes in morphology of four subtypes of GCs (ON-type RGA2 [ON-RGA2], OFF-type RGA2 [OFF-RGA2], ON-type RGC1 [ON-RGC1], and ON-OFF type RGD2 [ON-OFF RGD2]) were characterized in Thy1-YFP transgenic mice. Using whole-cell patch-clamp recording, passive membrane properties and action potential (AP) firing properties were further investigated in transient ON- and OFF-RGA2 cells. Results: Morphologic parameters were significantly altered in the dendrites branching in the ON sublamina of the inner plexiform layer (IPL) for ON-RGA2 cells and ON-OFF RGD2 cells. Much less significant changes, if any, were seen in those arborizing in the OFF sublamina of the IPL for OFF-RGA2 and ON-OFF RGD2 cells. No detectable changes in morphology were seen in RGC1 cells. Electrophysiologically, increased resting membrane potentials and decreased membrane capacitance were found in transient ON-RGA2 cells, but not in transient OFF-RGA2 cells. Similar alterations in AP firing properties, such as an increase in AP width and reduction in maximum spiking rate, were shared by these two subtypes. Furthermore, in response to depolarizing current injections, both cells generated more APs suggesting an enhanced excitability of these cells in diabetic conditions. Conclusions: These differential changes in morphology and electrophysiology in subtypes of GCs may be responsible for reduced contrast sensitivity known to occur during the early stage of diabetic retinopathy.


Asunto(s)
Diabetes Mellitus Experimental/patología , Retinopatía Diabética/patología , Células Ganglionares de la Retina/patología , Animales , Proteínas Bacterianas/metabolismo , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Retinopatía Diabética/sangre , Modelos Animales de Enfermedad , Femenino , Proteínas Luminiscentes/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Placa-Clamp , Estimulación Luminosa , Estreptozocina
18.
Neuropharmacology ; 133: 38-50, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29325900

RESUMEN

Orexin-A, -B play a crucial role in arousal and feeding by activating two G-protein-coupled receptors: orexin receptor 1 (OX1R) and orexin receptor 2 (OX2R). Orexins, along with orexin receptors, are expressed in retinal neurons, and they have been shown to differentially modulate excitatory AMPA receptors of amacrine and ganglion cells in the inner retina. In this work we report that orexin-B modulates the activity of rod bipolar cells (RBCs) located in the outer retina of rat. Intravitreal injection of orexin-B increased the amplitude of the scotopic electroretinographic b-wave, a reflection of RBC activity, recorded in vivo. Patch clamp recordings in rat retinal slices showed that orexin-B did not change glutamatergic excitatory component of the RBC response driven by photoreceptors. Effects of orexin-B on GABA receptor-mediated synaptic transmission of RBCs were then examined. In retinal slice preparations orexin-B suppressed GABA receptor-mediated inhibitory postsynaptic currents of RBCs in the inner plexiform layer. Furthermore, using whole-cell recordings in isolated RBCs it was shown that orexin-B suppressed GABAC receptor-, but not GABAA receptor-, mediated currents of the RBCs, an effect that was blocked by OX1R and OX2R antagonists. The orexin-B-induced inhibition of GABAC currents was likely mediated by a Gi/o/PC-PLC/Ca2+-independent PKC signaling pathway, as such inhibition was absent when each step of the above-pathway was blocked with GDP-ß-S/pertussis toxin (for Gi/o), D609 (for PLC), bisindolylmaleimide IV (for PKC)/rottlerin (for PKCδ), respectively. The orexin-B-induced potentiation of RBC activity may improve visual acuity and contrast sensitivity of the animal during the dark period (wake phase).


Asunto(s)
Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Orexinas/farmacología , Retina/citología , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Animales Recién Nacidos , Calcio/metabolismo , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , GABAérgicos/farmacología , Glicina/farmacología , Luz , Masculino , Potenciales de la Membrana/efectos de los fármacos , Receptores de Orexina/metabolismo , Técnicas de Placa-Clamp , Propionatos/farmacología , Ratas , Ratas Sprague-Dawley , Ácido gamma-Aminobutírico/farmacología
19.
Neurosci Lett ; 421(2): 106-9, 2007 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-17566658

RESUMEN

Natriuretic peptides (NPs) may act as neuromodulators through activation of three specific receptor subtypes (NPRs). In the present study we examined the expression of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP) on different subtypes of retinal amacrine cells (ACs) in rat by immunofluorescence double labeling. All three NPs were moderately expressed in dopaminergic and cholinergic ACs, stained by tyrosine hydroxylase (TH) and choline acetyltransferase (ChAT), respectively. The immunostaining appeared on the membrane, cytoplasm and somatodendritic compartments of these ACs. In AII glycinergic ACs, labeled by parvalbumin (PV), however, only faint punctate staining, if any, was seen. These results suggest that NPs could be produced in ACs and play a neuromodulatory role in the inner retina. Together with a previous immunocytochemical study, showing that NPR-B is present in cultured rat GABAergic ACs, our results further suggest that NPs produced in ACs may also modulate their own activity.


Asunto(s)
Células Amacrinas/metabolismo , Péptidos Natriuréticos/metabolismo , Retina/citología , Animales , Colina O-Acetiltransferasa/metabolismo , Inmunohistoquímica , Masculino , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa/metabolismo
20.
Brain Res Bull ; 73(4-6): 273-7, 2007 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-17562393

RESUMEN

Expression of 5-hydroxytryptamine (5-HT) 2A receptor (5-HT2A) was studied in bullfrog and rat retinas by immunocytochemistry. In the bullfrog retina, 5-HT2A-immunoreactivity was observed in both the outer and inner plexiform layers (OPL and IPL). Double labeling experiments further showed that 5-HT2A was expressed in Müller cells stained by GFAP. Labeling for 5-HT2A was strong in the somata and endfeet and relatively weak in the major processes and fine branchets of Müller cells. In contrast, 5-HT2A immunoreactivity was hardly detected in the rat retina, and no rat Müller cells were labeled. Furthermore, immunocytochemical assay demonstrated that labeling for 5-HT was present in amacrine cells and displaced amacrine cells in the inner retina of bullfrog, but not in the rat retina. These results suggest that 5-HT may modulate retinal information processing via activating 5-HT2A expressed in neuronal and glial elements in bullfrog, but that such modulation is unlikely to occur in rat.


Asunto(s)
Receptor de Serotonina 5-HT2A/metabolismo , Retina/citología , Animales , Inmunohistoquímica , Neuronas/citología , Neuronas/metabolismo , Rana catesbeiana , Ratas , Retina/metabolismo , Serotonina/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda