Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Neural Regen Res ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38934389

ABSTRACT

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.

2.
iScience ; 26(9): 107680, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37680468

ABSTRACT

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.

3.
Sci Adv ; 9(12): eadf4651, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36947616

ABSTRACT

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.


Subject(s)
Central Amygdaloid Nucleus , Retinal Ganglion Cells , Mice , Animals , Retinal Ganglion Cells/metabolism , Retina , Retinal Cone Photoreceptor Cells , Photoreceptor Cells, Vertebrate/metabolism , Light
4.
Curr Biol ; 33(7): 1372-1380.e4, 2023 04 10.
Article in English | MEDLINE | ID: mdl-36893758

ABSTRACT

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.


Subject(s)
Drosophila , Nociception , Animals , Neurons/physiology , Afferent Pathways , Nociceptors
5.
Prog Retin Eye Res ; 93: 101169, 2023 03.
Article in English | MEDLINE | ID: mdl-36736070

ABSTRACT

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.


Subject(s)
Glaucoma , Gliosis , Animals , Gliosis/pathology , Retina/metabolism , Retinal Ganglion Cells/pathology , Neuroglia/pathology , Intraocular Pressure , Disease Models, Animal
6.
Sci Adv ; 8(23): eabm9027, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35675393

ABSTRACT

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.


Subject(s)
Myopia , Retinal Ganglion Cells , Animals , Mice , Myopia/etiology , Photoreceptor Cells, Vertebrate , Retinal Ganglion Cells/physiology , Vision, Ocular
7.
Neurosci Bull ; 38(9): 992-1006, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35349094

ABSTRACT

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.


Subject(s)
Melatonin , Myopia , Animals , Disease Models, Animal , Dopamine , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Retina , Sensory Deprivation
8.
J Neuroinflammation ; 18(1): 303, 2021 Dec 24.
Article in English | MEDLINE | ID: mdl-34952606

ABSTRACT

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.


Subject(s)
Ependymoglial Cells/pathology , Glaucoma/complications , Microglia/pathology , Retinitis/etiology , Retinitis/pathology , Adenosine Triphosphate/physiology , Animals , Coculture Techniques , Cytokines/metabolism , Macrophage Activation , Mice , Mice, Inbred C57BL , Ocular Hypertension/complications , Receptors, Purinergic P2X7 , Retinal Ganglion Cells/pathology , Signal Transduction
9.
J Neurosci ; 41(12): 2566-2580, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33536197

ABSTRACT

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.


Subject(s)
Orexins/administration & dosage , Photic Stimulation/methods , Pupil/drug effects , Reflex, Pupillary/drug effects , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Animals , Benzimidazoles/administration & dosage , Female , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orexin Receptors/agonists , Orexin Receptors/metabolism , Orexins/antagonists & inhibitors , Pupil/physiology , Pyrrolidines/administration & dosage , Reflex, Pupillary/physiology , Retinal Ganglion Cells/metabolism
10.
Diabetes ; 70(5): 1157-1169, 2021 05.
Article in English | MEDLINE | ID: mdl-33574020

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Retina/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Streptozocin/toxicity , Animals , Mice , Retina/drug effects
11.
Cells ; 9(12)2020 12 01.
Article in English | MEDLINE | ID: mdl-33271919

ABSTRACT

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.


Subject(s)
Acetylcholine/metabolism , Action Potentials/physiology , Amacrine Cells/metabolism , Amacrine Cells/physiology , Glutamates/metabolism , Retina/physiology , Animals , Animals, Newborn , Calcium/metabolism , Cholinergic Agents/metabolism , Female , Male , Mice , Retina/metabolism
12.
Cell Death Dis ; 11(9): 734, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32913260

ABSTRACT

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.


Subject(s)
Ocular Hypertension/genetics , Peptide Fragments/metabolism , Retinal Ganglion Cells/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Apoptosis , Cell Differentiation , Chronic Disease , Disease Models, Animal , Humans , Male , Mice , Ocular Hypertension/pathology
13.
Invest Ophthalmol Vis Sci ; 61(2): 14, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32049344

ABSTRACT

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.


Subject(s)
Amacrine Cells/metabolism , Homeodomain Proteins/metabolism , Retinal Ganglion Cells/metabolism , Transcription Factor Brn-3B/metabolism , Transgenes/physiology , Animals , Channelrhodopsins/metabolism , Choline O-Acetyltransferase/metabolism , Female , Gene Expression , Male , Mice, Inbred C57BL , Mice, Transgenic , Transgenes/genetics
14.
Brain Res ; 1726: 146537, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31672473

ABSTRACT

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.


Subject(s)
Myopia/physiopathology , Optic Nerve/growth & development , Retina/growth & development , Visual Pathways/growth & development , 3,4-Dihydroxyphenylacetic Acid/metabolism , Amacrine Cells/metabolism , Animals , Cornea/growth & development , Cornea/pathology , Dopamine/metabolism , Mice, Inbred C57BL , Myopia/metabolism , Myopia/pathology , Nerve Crush , Retina/metabolism , Retina/pathology , Tyrosine 3-Monooxygenase/metabolism , Visual Pathways/metabolism
15.
Invest Ophthalmol Vis Sci ; 60(5): 1644-1656, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30995300

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/pathology , Retinal Ganglion Cells/pathology , Animals , Bacterial Proteins/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetic Retinopathy/blood , Disease Models, Animal , Female , Luminescent Proteins/metabolism , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques , Photic Stimulation , Streptozocin
16.
Neurosci Bull ; 35(4): 673-687, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30888607

ABSTRACT

Ras-related C3 botulinum toxin substrate 1 (Rac1), a member of the Rho GTPase family which plays important roles in dendritic spine morphology and plasticity, is a key regulator of cytoskeletal reorganization in dendrites and spines. Here, we investigated whether and how Rac1 modulates synaptic transmission in mouse retinal ganglion cells (RGCs) using selective conditional knockout of Rac1 (Rac1-cKO). Rac1-cKO significantly reduced the frequency of AMPA receptor-mediated miniature excitatory postsynaptic currents, while glycine/GABAA receptor-mediated miniature inhibitory postsynaptic currents were not affected. Although the total GluA1 protein level was increased in Rac1-cKO mice, its expression in the membrane component was unchanged. Rac1-cKO did not affect spine-like branch density in single dendrites, but significantly reduced the dendritic complexity, which resulted in a decrease in the total number of dendritic spine-like branches. These results suggest that Rac1 selectively affects excitatory synaptic transmission in RGCs by modulating dendritic complexity.


Subject(s)
Dendrites/metabolism , Neuropeptides/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Synaptic Transmission/genetics , rac1 GTP-Binding Protein/metabolism , Animals , Dendrites/ultrastructure , Dendritic Spines/metabolism , Excitatory Postsynaptic Potentials/physiology , GABA-A Receptor Antagonists , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neuropeptides/deficiency , Receptors, AMPA/metabolism , Receptors, GABA-A/metabolism , Receptors, Glycine/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Synapses/metabolism , rac1 GTP-Binding Protein/deficiency
17.
Acta Neuropathol Commun ; 6(1): 111, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30355282

ABSTRACT

It was previously shown that EphB/ephrinB reverse signaling in retinal ganglion cells (RGCs) is activated and involved in RGC apoptosis in a rat chronic ocular hypertension (COH) model. In the present work, we first show that ephrinB/EphB forward signaling was activated in COH retinas, and RGC apoptosis in COH retinas was reduced by PP2, an inhibitor of ephrinB/EphB forward signaling. We further demonstrate that treatment of cultured Müller cells with ephrinB1-Fc, an EphB1 activator, or intravitreal injection of ephrinB1-Fc in normal rats induced an increase in phosphorylated EphB levels in these cells, indicating the activation of ephrinB/EphB forward signaling, similar to those in COH retinas. The ephrinB1-Fc treatment did not induce Müller cell gliosis, as evidenced by unchanged GFAP expression, but significantly up-regulated mRNA and protein levels of tumor necrosis factor-α (TNF-α) in Müller cells, thereby promoting RGC apoptosis. Production of TNF-α induced by the activation of ephrinB/EphB forward signaling was mediated by the NR2B subunit of NMDA receptors, which was followed by a distinct PI3K/Akt/NF-κB signaling pathway, as pharmacological interference of each step of this pathway caused a reduction of TNF-α production, thus attenuating RGC apoptosis. Functional analysis of forward and reverse signaling in such a unique system, in which ephrin and Eph exist respectively in a glial element and a neuronal element, is of theoretical importance. Moreover, our results also raise a possibility that suppression of ephrinB/EphB forward signaling may be a new strategy for ameliorating RGC apoptosis in glaucoma.


Subject(s)
Apoptosis/physiology , Ephrin-B1/metabolism , Glaucoma/pathology , Receptors, Eph Family/metabolism , Retinal Ganglion Cells/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Animals, Newborn , Antioxidants/pharmacology , Apoptosis/drug effects , Cells, Cultured , Chromones/pharmacology , Disease Models, Animal , Ephrin-B1/pharmacology , Excitatory Amino Acid Agents/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Morpholines/pharmacology , Phenols/pharmacology , Piperidines/pharmacology , Proline/analogs & derivatives , Proline/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Eph Family/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Ganglion Cells/drug effects , Signal Transduction , Thiocarbamates/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
18.
Glia ; 66(7): 1507-1519, 2018 07.
Article in English | MEDLINE | ID: mdl-29508439

ABSTRACT

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.


Subject(s)
Ependymoglial Cells/metabolism , Gliosis/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Ocular Hypertension/metabolism , Receptors, Dopamine D1/metabolism , Animals , Disease Models, Animal , Ependymoglial Cells/pathology , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Male , Membrane Potentials/physiology , Ocular Hypertension/pathology , Rats, Sprague-Dawley , Receptors, Dopamine D1/antagonists & inhibitors , Tyrosine 3-Monooxygenase/metabolism , Vitreous Body/metabolism , Vitreous Body/pathology
19.
Methods Mol Biol ; 1753: 289-303, 2018.
Article in English | MEDLINE | ID: mdl-29564797

ABSTRACT

Intrinsically photosensitive retinal ganglion cells (ipRGCs) are a special subset of retinal output neurons capable of detecting and responding to light via a unique photopigment called melanopsin. Melanopsin activation is essential to a wide array of physiological functions, especially to those related to non-image-forming vision. Since ipRGCs only constitute a very small proportion of retinal ganglion cells, targeted recording of melanopsin-driven responses used to be a big challenge to vision researchers. Multielectrode array (MEA) recording provides a noninvasive, high throughput method to monitor melanopsin-driven responses. When synaptic inputs from rod/cone photoreceptors are silenced with glutamatergic blockers, extracellular electric signals derived from melanopsin activation can be recorded from multiple ipRGCs simultaneously by tens of microelectrodes aligned in an array. In this chapter we describe how our labs have approached MEA recording of melanopsin-driven light responses in adult mouse retinas. Instruments, tools and chemical reagents routinely used for setting up a successful MEA recording are listed, and a standard experimental procedure is provided. The implementation of this technique offers a useful paradigm that can be used to conduct functional assessments of ipRGCs and NIF vision.


Subject(s)
Patch-Clamp Techniques/methods , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Vision, Ocular/physiology , Animals , Mice , Mice, Inbred C57BL , Microelectrodes , Patch-Clamp Techniques/instrumentation , Photic Stimulation/instrumentation , Photic Stimulation/methods , Visual Pathways/metabolism
20.
Neuropharmacology ; 133: 38-50, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29325900

ABSTRACT

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).


Subject(s)
Inhibitory Postsynaptic Potentials/drug effects , Orexins/pharmacology , Retina/cytology , Retinal Rod Photoreceptor Cells/drug effects , Synaptic Transmission/drug effects , Animals , Animals, Newborn , Calcium/metabolism , Excitatory Amino Acid Agents/pharmacology , GABA Agents/pharmacology , Glycine/pharmacology , Light , Male , Membrane Potentials/drug effects , Orexin Receptors/metabolism , Patch-Clamp Techniques , Propionates/pharmacology , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...