Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 80
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.
J Neuroinflammation ; 18(1): 303, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34952606

RESUMEN

BACKGROUND: Glaucoma, the leading cause of irreversible blindness, is a retinal neurodegenerative disease, which results from progressive apoptotic death of retinal ganglion cells (RGCs). Although the mechanisms underlying RGC apoptosis in glaucoma are extremely complicated, an abnormal cross-talk between retinal glial cells and RGCs is generally thought to be involved. However, how interaction of Müller cells and microglia, two types of glial cells, contributes to RGC injury is largely unknown. METHODS: A mouse chronic ocular hypertension (COH) experimental glaucoma model was produced. Western blotting, immunofluorescence, quantitative real-time polymerase chain reaction (q-PCR), transwell co-culture of glial cells, flow cytometry assay, ELISA, Ca2+ image, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) techniques were employed to investigate the interaction of Müller cells and microglia, and its underlying mechanisms in COH retina. RESULTS: We first showed that Müller cell activation in mice with COH induced microglia activation through the ATP/P2X7 receptor pathway. The activation of microglia resulted in a significant increase in mRNA and protein levels of pro-inflammatory factors, such as tumor necrosis factor-α and interleukin-6. These inflammatory factors in turn caused the up-regulation of mRNA expression of pro-inflammatory factors in Müller cells through a positive feedback manner. CONCLUSIONS: These findings provide robust evidence, for the first time, that retinal inflammatory response may be aggravated by an interplay between activated two types of glial cells. These results also suggest that to reduce the interplay between Müller cells and microglia could be a potential effective strategy for preventing the loss of RGCs in glaucoma.


Asunto(s)
Células Ependimogliales/patología , Glaucoma/complicaciones , Microglía/patología , Retinitis/etiología , Retinitis/patología , Adenosina Trifosfato/fisiología , Animales , Técnicas de Cocultivo , Citocinas/metabolismo , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Hipertensión Ocular/complicaciones , Receptores Purinérgicos P2X7 , Células Ganglionares de la Retina/patología , Transducción de Señal
3.
Glia ; 66(7): 1507-1519, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29508439

RESUMEN

Müller cell gliosis is a common response in many retinal pathological conditions. We previously demonstrated that downregulation of Kir channels contributes to Müller cell gliosis in a rat chronic ocular hypertension (COH) model. Here, the possible involvement of outward K+ currents in Müller cell gliosis was investigated. Outward K+ current densities in Müller cells isolated from COH rats, as compared with those in normal rats, showed a significant increase, which was mainly contributed by large-conductance Ca2+ -activated K+ (BKCa ) channels. The involvement of BKCa channels in Müller cell gliosis is suggested by the fact that glial fibrillary acidic protein (GFAP) levels were augmented in COH retinas when these channels were suppressed by intravitreal injections of iberiotoxin. In COH retinas an increase in dopamine (DA) D1 receptor (D1R) expression in Müller cells was revealed by both immunohistochemistry and Western blotting. Moreover, protein levels of tyrosine hydroxylase were also increased, and consistent to this, retinal DA contents were elevated. SKF81297, a selective D1R agonist, enhanced BKCa currents of normal Müller cells through intracellular cAMP-PKA signaling pathway. Furthermore, GFAP levels were increased by the D1R antagonist SCH23390 injected intravitreally through eliminating the BKCa current upregulation in COH retinas, but partially reduced by SKF81297. All these results strongly suggest that the DA-D1R system may be activated to a stronger extent in COH rat retinas, thus increasing BKCa currents of Müller cells. The upregulation of BKCa channels may antagonize the Kir channel inhibition-induced depolarization of Müller cells, thereby attenuating the gliosis of these cells.


Asunto(s)
Células Ependimogliales/metabolismo , Gliosis/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Hipertensión Ocular/metabolismo , Receptores de Dopamina D1/metabolismo , Animales , Modelos Animales de Enfermedad , Células Ependimogliales/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/patología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/antagonistas & inhibidores , Masculino , Potenciales de la Membrana/fisiología , Hipertensión Ocular/patología , Ratas Sprague-Dawley , Receptores de Dopamina D1/antagonistas & inhibidores , Tirosina 3-Monooxigenasa/metabolismo , Cuerpo Vítreo/metabolismo , Cuerpo Vítreo/patología
4.
J Neurosci ; 35(13): 5409-21, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25834064

RESUMEN

EphB1, expressed in Müller cells, and ephrinB2, expressed in both Müller cells and retinal ganglion cells (RGCs), constitute an EphB/ephrinB reverse signaling in RGCs. Whether and how this reverse signaling is involved in RGC apoptosis in a rat chronic ocular hypertension (COH) model was investigated. In the COH model, both EphB1 and ephrinB2 were significantly increased and the reverse signaling was activated, which was accompanied by increased protein levels of phosphorylated (p) src, GluA2, and p-GluA2. Intravitreal injection of EphB2-Fc, an activator of ephrinB2, induced an increase in TUNEL-positive signals in normal retinae. A coimmunoprecipitation assay demonstrated direct interactions among ephrinB2, p-src, and GluA2. Moreover, in COH rats the expression of GluA2 proteins on the surface of retinal cells was decreased. Such GluA2 endocytosis could be prevented by preoperational intravitreal injection of 4-amino-3-(4-chlorophenyl)-1-(t-butyl)-1H-pyrazolo [3,4-d] pyrimidine (PP2), an inhibitor of src family tyrosine kinases, and possibly involved the protein interacting with C kinase 1 and phosphorylation of GluA2. In normal rats, intravitreal injection of EphB2-Fc caused changes in these protein levels similar to those observed in COH rats, which all could be avoided by preinjection of PP2. Patch-clamp experiments further showed that the current-voltage relationship of AMPA receptor-mediated EPSCs of RGCs exhibited stronger inward rectification in EphB2-Fc-injected rats. Furthermore, preinjection of PP2 or N-[3-[[4-[(3-aminopropyl)amino]butyl]amino]propyl]-1-naphthaleneacetamide trihydrochloride) (Naspm), a Ca(2+)-permeable GluA2-lacking AMPA receptor inhibitor, remarkably inhibited RGC apoptosis in either EphB2-Fc-injected or COH rats. Together, elevated GluA2 trafficking induced by activated EphB2/ephrinB2 reverse signaling likely contributes to RGC apoptosis in COH rats.


Asunto(s)
Apoptosis/fisiología , Efrina-B2/metabolismo , Hipertensión Ocular/metabolismo , Receptor EphB1/metabolismo , Receptores AMPA/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/fisiología , Transducción de Señal , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Efrina-B2/agonistas , Potenciales Postsinápticos Excitadores , Etiquetado Corte-Fin in Situ , Masculino , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Pirimidinas/farmacología , Ratas , Receptores AMPA/antagonistas & inhibidores , Células Ganglionares de la Retina/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
5.
Sheng Li Xue Bao ; 68(4): 483-91, 2016 Aug 25.
Artículo en Zh | MEDLINE | ID: mdl-27546508

RESUMEN

Glaucoma, the second leading cause of blindness, is a neurodegenerative disease characterized by optic nerve degeneration related to apoptotic death of retinal ganglion cells (RGCs). In the pathogenesis of RGC death following the onset of glaucoma, functional changes of glutamate receptors are commonly regarded as important risk factors. During the past several years, we have explored the mechanisms underlying RGC apoptosis and retinal Müller cell reactivation (gliosis) in a rat chronic ocular hypertension (COH) model. We demonstrated that elevated intraocular pressure in COH rats may induce changes of various signaling pathways, which are involved in RGC apoptosis by modulating glutamate NMDA and AMPA receptors. Moreover, we also demonstrated that over-activation of group I metabotropic glutamate receptors (mGluR I) by excessive extracellular glutamate in COH rats could contribute to Müller cell gliosis by suppressing Kir4.1 channels. In this review, incorporating our results, we discuss glutamate receptor- mediated RGC apoptosis and Müller cell gliosis in experimental glaucoma.


Asunto(s)
Glaucoma , Retina , Animales , Modelos Animales de Enfermedad , Hipertensión Ocular , Receptores de Glutamato , Células Ganglionares de la Retina
6.
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.

7.
J Neurosci ; 32(37): 12744-55, 2012 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-22972998

RESUMEN

Müller cell gliosis, which is characterized by upregulated expression of glial fibrillary acidic protein (GFAP), is a universal response in many retinal pathological conditions. Whether down-regulation of inward rectifying K+ (Kir) channels, which commonly accompanies the enhanced GFAP expression, could contribute to Müller cell gliosis is poorly understood. We investigated changes of Kir currents, GFAP and Kir4.1 protein expression in Müller cells in a rat chronic ocular hypertension (COH) model, and explored the mechanisms underlying Müller cell gliosis. We show that Kir currents and Kir4.1 protein expression in Müller cells were reduced significantly, while GFAP expression was increased in COH rats, and these changes were eliminated by MPEP, a group I metabotropic glutamate receptors (mGluR I) subtype mGluR5 antagonist. In normal isolated Müller cells, the mGluR I agonist (S)-3,5-dihydroxyphenylglycine (DHPG) suppressed the Kir currents and the suppression was blocked by MPEP. The DHPG effect was mediated by the intracellular Ca2+ -dependent PLC/IP3-ryanodine/PKC signaling pathway, but the cAMP-PKA pathway was not involved. Moreover, intravitreal injection of DHPG in normal rats induced changes in Müller cells, similar to those observed in COH rats. The DHPG-induced increase of GFAP expression in Müller cells was obstructed by Ba2+, suggesting the involvement of Kir channels. We conclude that overactivation of mGluR5 by excessive extracellular glutamate in COH rats could contribute to Müller cell gliosis by suppressing Kir channels.


Asunto(s)
Modelos Animales de Enfermedad , Gliosis/fisiopatología , Hipertensión Ocular/fisiopatología , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Enfermedades de la Retina/fisiopatología , Animales , Enfermedad Crónica , Gliosis/etiología , Humanos , Activación del Canal Iónico , Masculino , Hipertensión Ocular/complicaciones , Ratas , Ratas Sprague-Dawley , Enfermedades de la Retina/etiología
8.
Prog Retin Eye Res ; 93: 101169, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36736070

RESUMEN

Elevation of intraocular pressure (IOP) is a major risk factor for neurodegeneration in glaucoma. Glial cells, which play an important role in normal functioning of retinal neurons, are well involved into retinal ganglion cell (RGC) degeneration in experimental glaucoma animal models generated by elevated IOP. In response to elevated IOP, mGluR I is first activated and Kir4.1 channels are subsequently inhibited, which leads to the activation of Müller cells. Müller cell activation is followed by a complex process, including proliferation, release of inflammatory and growth factors (gliosis). Gliosis is further regulated by several factors. Activated Müller cells contribute to RGC degeneration through generating glutamate receptor-mediated excitotoxicity, releasing cytotoxic factors and inducing microglia activation. Elevated IOP activates microglia, and following morphological and functional changes, these cells, as resident immune cells in the retina, show adaptive immune responses, including an enhanced release of pro-inflammatory factors (tumor neurosis factor-α, interleukins, etc.). These ATP and Toll-like receptor-mediated responses are further regulated by heat shock proteins, CD200R, chemokine receptors, and metabotropic purinergic receptors, may aggravate RGC loss. In the optic nerve head, astrogliosis is initiated and regulated by a complex reaction process, including purines, transmitters, chemokines, growth factors and cytokines, which contributes to RGC axon injury through releasing pro-inflammatory factors and changing extracellular matrix in glaucoma. The effects of activated glial cells on RGCs are further modified by the interplay among different types of glial cells. This review is concluded by presenting an in-depth discussion of possible research directions in this field in the future.


Asunto(s)
Glaucoma , Gliosis , Animales , Gliosis/patología , Retina/metabolismo , Células Ganglionares de la Retina/patología , Neuroglía/patología , Presión Intraocular , Modelos Animales de Enfermedad
9.
Curr Biol ; 33(7): 1372-1380.e4, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-36893758

RESUMEN

Animals' response to a stimulus in one sensory modality is usually influenced by other modalities.1 One important type of multisensory integration is the cross-modal modulation, in which one sensory modality modulates (typically inhibits) another. Identification of the mechanisms underlying cross-modal modulations is crucial for understanding how sensory inputs shape animals' perception and for understanding sensory processing disorders.2,3,4 However, the synaptic and circuit mechanisms that underlie cross-modal modulation are poorly understood. This is due to the difficulty of separating cross-modal modulation from multisensory integrations in neurons that receive excitatory inputs from two or more sensory modalities5-in which case it is unclear what the modulating or modulated modality is. In this study, we report a unique system for studying cross-modal modulation by taking advantage of the genetic resources in Drosophila. We show that gentle mechanical stimuli inhibit nociceptive responses in Drosophila larvae. Low-threshold mechanosensory neurons inhibit a key second-order neuron in the nociceptive pathway through metabotropic GABA receptors on nociceptor synaptic terminals. Strikingly, this cross-modal inhibition is only effective when nociceptor inputs are weak, thus serving as a gating mechanism for filtering out weak nociceptive inputs. Our findings unveil a novel cross-modal gating mechanism for sensory pathways.


Asunto(s)
Drosophila , Nocicepción , Animales , Neuronas/fisiología , Vías Aferentes , Nociceptores
10.
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.

11.
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
12.
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
13.
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
14.
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
15.
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
16.
J Physiol ; 588(Pt 14): 2605-19, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20519319

RESUMEN

In vertebrate retina, melatonin regulates various physiological functions. In this work we investigated the mechanisms underlying melatonin-induced potentiation of glycine currents in rat retinal ganglion cells (RGCs). Immunofluorescence double labelling showed that rat RGCs were solely immunoreactive to melatonin MT(2) receptors. Melatonin potentiated glycine currents of RGCs, which was reversed by the MT(2) receptor antagonist 4-P-PDOT. The melatonin effect was blocked by intracellular dialysis of GDP-beta-S. Either preincubation with pertussis toxin or application of the phosphatidylcholine (PC)-specific phospholipase C (PLC) inhibitor D609, but not the phosphatidylinositol (PI)-PLC inhibitor U73122, blocked the melatonin effect. The protein kinase C (PKC) activator PMA potentiated the glycine currents and in the presence of PMA melatonin failed to cause further potentiation of the currents, whereas application of the PKC inhibitor bisindolylmaleimide IV abolished the melatonin-induced potentiation. The melatonin effect persisted when [Ca(2+)](i) was chelated by BAPTA, and melatonin induced no increase in [Ca(2+)](i). Neither cAMP-PKA nor cGMP-PKG signalling pathways seemed to be involved because 8-Br-cAMP or 8-Br-cGMP failed to cause potentiation of the glycine currents and both the PKA inhibitor H-89 and the PKG inhibitor KT5823 did not block the melatonin-induced potentiation. In consequence, a distinct PC-PLC/PKC signalling pathway, following the activation of G(i/o)-coupled MT(2) receptors, is most likely responsible for the melatonin-induced potentiation of glycine currents of rat RGCs. Furthermore, in rat retinal slices melatonin potentiated light-evoked glycine receptor-mediated inhibitory postsynaptic currents in RGCs. These results suggest that melatonin, being at higher levels at night, may help animals to detect positive or negative contrast in night vision by modulating inhibitory signals largely mediated by glycinergic amacrine cells in the inner retina.


Asunto(s)
Melatonina/fisiología , Proteína Quinasa C/fisiología , Células Ganglionares de la Retina/fisiología , Fosfolipasas de Tipo C/fisiología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Hidrocarburos Aromáticos con Puentes/farmacología , Calcio/análisis , Carbazoles/farmacología , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Estrenos/farmacología , Glicina/fisiología , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Indoles/farmacología , Isoquinolinas/farmacología , Masculino , Maleimidas/farmacología , Norbornanos , Toxina del Pertussis/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Pirrolidinonas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Melatonina MT2/antagonistas & inhibidores , Receptor de Melatonina MT2/fisiología , Células Ganglionares de la Retina/enzimología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Acetato de Tetradecanoilforbol/farmacología , Tetrahidronaftalenos/farmacología , Tiocarbamatos , Tionas/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores
17.
Prog Neurobiol ; 84(3): 234-48, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18215455

RESUMEN

Natriuretic peptides (NPs), including atrial, brain and C-type NPs, are a family of structurally related but genetically distinct peptides. These peptides, along with their receptors (NPRs), are long known to be involved in the regulation of various physiological functions, such as diuresis, natriuresis, and blood flow. Recently, abundant evidence shows that NPs and NPRs are widely distributed in the central nervous system (CNS), suggesting possible roles of NPs in modulating physiological functions of the CNS. This review starts with a brief summary of relevant background information, such as molecular structures of NPs and NPRs and general intracellular mechanisms after activation of NPRs. We then provide a detailed description of the expression profiles of NPs and NPRs in the CNS and an in-depth discussion of how NPs are involved in neural development, neurotransmitter release, synaptic transmission and neuroprotection through activation of NPRs.


Asunto(s)
Factor Natriurético Atrial/fisiología , Sistema Nervioso Central/fisiología , Péptidos Natriuréticos/metabolismo , Animales , Humanos
18.
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
19.
Cell Death Dis ; 11(9): 734, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32913260

RESUMEN

Autophagy has a fundamental role in maintaining cell homeostasis. Although autophagy has been implicated in glaucomatous pathology, how it regulates retinal ganglion cell (RGC) injury is largely unknown. In the present work, we found that biphasic autophagy in RGCs occurred in a mouse model of chronic ocular hypertension (COH), accompanied by activation of Rac1, a member of the Rho family. Rac1 conditional knockout (Rac1 cKO) in RGCs attenuated RGC apoptosis, in addition to blocking the increase in the number of autophagosomes and the expression of autophagy-related proteins (Beclin1, LC3-II/I, and p62) in COH retinas. Electron micrograph and double immunostaining of LAMP1 and LC3B showed that Rac1 cKO accelerated autolysosome fusion in RGC axons of COH mice. Inhibiting the first autophagic peak with 3-methyladenine or Atg13 siRNA reduced RGC apoptosis, whereas inhibiting the second autophagic peak with 3-MA or blocking autophagic flux by chloroquine increased RGC apoptosis. Furthermore, Rac1 cKO reduced the number of autophagosomes and apoptotic RGCs induced by rapamycin injected intravitreally, which suggests that Rac1 negatively regulates mTOR activity. Moreover, Rac1 deletion decreased Bak expression and did not interfere with the interaction of Beclin1 and Bcl-2 or Bak in COH retinas. In conclusion, autophagy promotes RGC apoptosis in the early stages of glaucoma and results in autophagic cell death in later stages. Rac1 deletion alleviates RGC damage by regulating the cross talk between autophagy and apoptosis through mTOR/Beclin1-Bak. Interfering with the Rac1/mTOR signaling pathway may provide a new strategy for treating glaucoma.


Asunto(s)
Hipertensión Ocular/genética , Fragmentos de Péptidos/metabolismo , Células Ganglionares de la Retina/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Apoptosis , Diferenciación Celular , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Hipertensión Ocular/patología
20.
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
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda