Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.550
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(11): 2767-2784.e23, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38733989

RESUMEN

The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.


Asunto(s)
Cerebelo , Neuronas , Retina , Animales , Femenino , Masculino , Ratones , Cerebelo/metabolismo , Cerebelo/irrigación sanguínea , Cerebelo/citología , Canales Iónicos/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Vasos Retinianos/metabolismo
2.
Cell ; 181(3): 590-603.e16, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32272060

RESUMEN

Conversion of glial cells into functional neurons represents a potential therapeutic approach for replenishing neuronal loss associated with neurodegenerative diseases and brain injury. Previous attempts in this area using expression of transcription factors were hindered by the low conversion efficiency and failure of generating desired neuronal types in vivo. Here, we report that downregulation of a single RNA-binding protein, polypyrimidine tract-binding protein 1 (Ptbp1), using in vivo viral delivery of a recently developed RNA-targeting CRISPR system CasRx, resulted in the conversion of Müller glia into retinal ganglion cells (RGCs) with a high efficiency, leading to the alleviation of disease symptoms associated with RGC loss. Furthermore, this approach also induced neurons with dopaminergic features in the striatum and alleviated motor defects in a Parkinson's disease mouse model. Thus, glia-to-neuron conversion by CasRx-mediated Ptbp1 knockdown represents a promising in vivo genetic approach for treating a variety of disorders due to neuronal loss.


Asunto(s)
Neurogénesis/fisiología , Neuroglía/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Sistemas CRISPR-Cas/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Modelos Animales de Enfermedad , Dopamina/metabolismo , Regulación de la Expresión Génica/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Células Ganglionares de la Retina/fisiología
3.
Cell ; 175(3): 652-664.e12, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30270038

RESUMEN

Non-image-forming vision in mammals is mediated primarily by melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs). In mouse M1-ipRGCs, by far the best-studied subtype, melanopsin activates PLCß4 (phospholipase C-ß4) to open TRPC6,7 channels, mechanistically similar to phototransduction in fly rhabdomeric (microvillous) photoreceptors. We report here that, surprisingly, mouse M4-ipRGCs rely on a different and hitherto undescribed melanopsin-driven, ciliary phototransduction mechanism involving cyclic nucleotide as the second messenger and HCN channels rather than CNG channels as the ion channel for phototransduction. Even more surprisingly, within an individual mouse M2-ipRGC, this HCN-channel-dependent, ciliary phototransduction pathway operates in parallel with the TRPC6,7-dependent rhabdomeric pathway. These findings reveal a complex heterogeneity in phototransduction among ipRGCs and, more importantly, break a general dogma about segregation of the two phototransduction motifs, likely with strong evolutionary implications.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Células Ganglionares de la Retina/metabolismo , Visión Ocular , Animales , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Nucleótidos Cíclicos/metabolismo , Células Ganglionares de la Retina/fisiología , Canales Catiónicos TRPC/metabolismo
4.
Annu Rev Neurosci ; 46: 1-15, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-36750409

RESUMEN

A holy grail of regenerative medicine is to replenish the cells that are lost due to disease. The adult mammalian central nervous system (CNS) has, however, largely lost such a regenerative ability. An emerging strategy for the generation of new neurons is through glia-to-neuron (GtN) conversion in vivo, mainly accomplished by the regulation of fate-determining factors. When inhibited, PTBP1, a factor involved in RNA biology, was reported to induce rapid and efficient GtN conversion in multiple regions of the adult CNS. Remarkably, PTBP1 inhibition was also claimed to greatly improve behaviors of mice with neurological diseases or aging. These phenomenal claims, if confirmed, would constitute a significant advancement in regenerative medicine. Unfortunately, neither GtN conversion nor therapeutic potential via PTBP1 inhibition was validated by the results of multiple subsequent replication studies with stringent methods. Here we review these controversial studies and conclude with recommendations for examining GtN conversion in vivo and future investigations of PTBP1.


Asunto(s)
Neuroglía , Neuronas , Animales , Ratones , Neuronas/fisiología , Sistema Nervioso Central , Retina , Mamíferos
5.
Proc Natl Acad Sci U S A ; 121(12): e2317218121, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38483997

RESUMEN

Across the animal kingdom, visual predation relies on motion-sensing neurons in the superior colliculus (SC) and its orthologs. These neurons exhibit complex stimulus preferences, including direction selectivity, which is thought to be critical for tracking the unpredictable escape routes of prey. The source of direction selectivity in the SC is contested, and its contributions to predation have not been tested experimentally. Here, we use type-specific cell removal to show that narrow-field (NF) neurons in the mouse SC guide predation. In vivo recordings demonstrate that direction-selective responses of NF cells are independent of recently reported stimulus-edge effects. Monosynaptic retrograde tracing reveals that NF cells receive synaptic input from direction-selective ganglion cells. When we eliminate direction selectivity in the retina of adult mice, direction-selective responses in the SC, including in NF cells, are lost. However, eliminating retinal direction selectivity does not affect the hunting success or strategies of mice, even when direction selectivity is removed after mice have learned to hunt, and despite abolishing the gaze-stabilizing optokinetic reflex. Thus, our results identify the retinal source of direction selectivity in the SC. They show that NF cells in the SC guide predation, an essential spatial orienting task, independent of their direction selectivity, revealing behavioral multiplexing of complex neural feature preferences and highlighting the importance of feature-selective manipulations for neuroethology.


Asunto(s)
Neuronas , Conducta Predatoria , Ratones , Animales , Neuronas/fisiología , Colículos Superiores/fisiología , Retina , Vías Visuales/fisiología
6.
Am J Hum Genet ; 110(3): 531-547, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36809767

RESUMEN

Familial dysautonomia (FD) is a rare neurodegenerative disease caused by a splicing mutation in elongator acetyltransferase complex subunit 1 (ELP1). This mutation leads to the skipping of exon 20 and a tissue-specific reduction of ELP1, mainly in the central and peripheral nervous systems. FD is a complex neurological disorder accompanied by severe gait ataxia and retinal degeneration. There is currently no effective treatment to restore ELP1 production in individuals with FD, and the disease is ultimately fatal. After identifying kinetin as a small molecule able to correct the ELP1 splicing defect, we worked on its optimization to generate novel splicing modulator compounds (SMCs) that can be used in individuals with FD. Here, we optimize the potency, efficacy, and bio-distribution of second-generation kinetin derivatives to develop an oral treatment for FD that can efficiently pass the blood-brain barrier and correct the ELP1 splicing defect in the nervous system. We demonstrate that the novel compound PTC258 efficiently restores correct ELP1 splicing in mouse tissues, including brain, and most importantly, prevents the progressive neuronal degeneration that is characteristic of FD. Postnatal oral administration of PTC258 to the phenotypic mouse model TgFD9;Elp1Δ20/flox increases full-length ELP1 transcript in a dose-dependent manner and leads to a 2-fold increase in functional ELP1 in the brain. Remarkably, PTC258 treatment improves survival, gait ataxia, and retinal degeneration in the phenotypic FD mice. Our findings highlight the great therapeutic potential of this novel class of small molecules as an oral treatment for FD.


Asunto(s)
Disautonomía Familiar , Enfermedades Neurodegenerativas , Degeneración Retiniana , Ratones , Animales , Disautonomía Familiar/genética , Cinetina , Ataxia de la Marcha , Administración Oral
7.
J Neurosci ; 44(5)2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38123991

RESUMEN

Maintaining precise synaptic contacts between neuronal partners is critical to ensure the proper functioning of the mammalian central nervous system (CNS). Diverse cell recognition molecules, such as classic cadherins (Cdhs), are part of the molecular machinery mediating synaptic choices during development and synaptic maintenance. Yet, the principles governing neuron-neuron wiring across diverse CNS neuron types remain largely unknown. The retinotectal synapses, connections from the retinal ganglion cells (RGCs) to the superior collicular (SC) neurons, offer an ideal experimental system to reveal molecular logic underlying synaptic choices and formation. This is due to the retina's unidirectional and laminar-restricted projections to the SC and the large databases of presynaptic RGC subtypes and postsynaptic SC neuronal types. Here, we focused on determining the role of Type II Cdhs in wiring the retinotectal synapses. We surveyed Cdhs expression patterns at neuronal resolution and revealed that Cdh13 is enriched in the wide-field neurons in the superficial SC (sSC). In either the Cdh13 null mutant or selective adult deletion within the wide-field neurons, there is a significant reduction of spine densities in the distal dendrites of these neurons in both sexes. Additionally, Cdh13 removal from presynaptic RGCs reduced dendritic spines in the postsynaptic wide-field neurons. Cdh13-expressing RGCs use differential mechanisms than αRGCs and On-Off Direction-Selective Ganglion Cells (ooDSGCs) to form specific retinotectal synapses. The results revealed a selective transneuronal interaction mediated by Cdh13 to maintain proper retinotectal synapses in vivo.


Asunto(s)
Células Ganglionares de la Retina , Sinapsis , Animales , Células Ganglionares de la Retina/fisiología , Sinapsis/fisiología , Colículos Superiores/fisiología , Dendritas/fisiología , Cadherinas/genética , Cadherinas/metabolismo , Mamíferos
8.
EMBO Rep ; 24(10): e56839, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37531065

RESUMEN

The formation of social memory between individuals of the opposite sex is crucial for expanding mating options or establishing monogamous pair bonding. A specialized neuronal circuit that regulates social memory could enhance an individual's mating opportunities and provide a parallel pathway for computing social behaviors. While the influence of light exposure on various forms of memory, such as fear and object memory, has been studied, its modulation of social recognition memory remains unclear. Here, we demonstrate that acute exposure to light impairs social recognition memory (SRM) in mice. Unlike sound and touch stimuli, light inhibits oxytocin neurons in the supraoptic nucleus (SON) via M1 SON-projecting intrinsically photosensitive retinal ganglion cells (ipRGCs) and GABAergic neurons in the perinuclear zone of the SON (pSON). We further show that optogenetic activation of SON oxytocin neurons using channelrhodopsin is sufficient to enhance SRM performance, even under light conditions. Our findings unveil a dedicated neuronal circuit through which luminance affects SRM, utilizing a non-image-forming visual pathway, distinct from the canonical modulatory role of the oxytocin system.

9.
Mol Ther ; 32(6): 1760-1778, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38659223

RESUMEN

Glaucoma is characterized by the progressive degeneration of retinal ganglion cells (RGCs) and their axons, and its risk increases with aging. Yet comprehensive insights into the complex mechanisms are largely unknown. Here, we found that anti-aging molecule Sirt6 was highly expressed in RGCs. Deleting Sirt6 globally or specifically in RGCs led to progressive RGC loss and optic nerve degeneration during aging, despite normal intraocular pressure (IOP), resembling a phenotype of normal-tension glaucoma. These detrimental effects were potentially mediated by accelerated RGC senescence through Caveolin-1 upregulation and by the induction of mitochondrial dysfunction. In mouse models of high-tension glaucoma, Sirt6 level was decreased after IOP elevation. Genetic overexpression of Sirt6 globally or specifically in RGCs significantly attenuated high tension-induced degeneration of RGCs and their axons, whereas partial or RGC-specific Sirt6 deletion accelerated RGC loss. Importantly, therapeutically targeting Sirt6 with pharmacological activator or AAV2-mediated gene delivery ameliorated high IOP-induced RGC degeneration. Together, our studies reveal a critical role of Sirt6 in preventing RGC and optic nerve degeneration during aging and glaucoma, setting the stage for further exploration of Sirt6 activation as a potential therapy for glaucoma.


Asunto(s)
Envejecimiento , Modelos Animales de Enfermedad , Glaucoma , Nervio Óptico , Células Ganglionares de la Retina , Sirtuinas , Animales , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Ratones , Sirtuinas/metabolismo , Sirtuinas/genética , Glaucoma/metabolismo , Glaucoma/genética , Glaucoma/patología , Glaucoma/etiología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Envejecimiento/metabolismo , Envejecimiento/genética , Presión Intraocular , Humanos , Axones/metabolismo , Axones/patología , Ratones Noqueados , Degeneración Nerviosa/metabolismo
10.
Genomics ; 116(1): 110776, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38163571

RESUMEN

The death of retinal ganglion cells (RGCs) can cause irreversible injury in visual function. Clarifying the mechanism of RGC degeneration is critical for the development of therapeutic strategies. Circular RNAs (circRNAs) are important regulators in many biological and pathological processes. Herein, we performed circRNA microarrays to identify dysregulated circRNAs following optic nerve crush (ONC). The results showed that 221 circRNAs were differentially expressed between ONC retinas and normal retinas. Notably, the levels of circular RNA-Dcaf6 (cDcaf6) expression in aqueous humor of glaucoma patients were higher than that in cataract patients. cDcaf6 silencing could reduce oxidative stress-induced RGC apoptosis in vitro and alleviate retinal neurodegeneration in vivo as shown by increased neuronal nuclei antigen (NeuN, neuronal bodies) and beta-III-tubulin (TUBB3, neuronal filaments) staining and reduced glial fibrillary acidic protein (GFAP, activated glial cells) and vimentin (activated glial cells) staining. Collectively, this study identifies a promising target for treating retinal neurodegeneration.


Asunto(s)
Traumatismos del Nervio Óptico , ARN Circular , Animales , Humanos , Modelos Animales de Enfermedad , Nervio Óptico/metabolismo , Nervio Óptico/patología , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/metabolismo , Retina , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , ARN Circular/genética , ARN Circular/metabolismo
11.
J Neurosci ; 43(29): 5319-5339, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37339877

RESUMEN

Saccades are a fundamental part of natural vision. They interrupt fixations of the visual gaze and rapidly shift the image that falls onto the retina. These stimulus dynamics can cause activation or suppression of different retinal ganglion cells, but how they affect the encoding of visual information in different types of ganglion cells is largely unknown. Here, we recorded spiking responses to saccade-like shifts of luminance gratings from ganglion cells in isolated marmoset retinas and investigated how the activity depended on the combination of presaccadic and postsaccadic images. All identified cell types, On and Off parasol and midget cells, as well as a type of Large Off cells, displayed distinct response patterns, including particular sensitivity to either the presaccadic or the postsaccadic image or combinations thereof. In addition, Off parasol and Large Off cells, but not On cells, showed pronounced sensitivity to whether the image changed across the transition. Stimulus sensitivity of On cells could be explained based on their responses to step changes in light intensity, whereas Off cells, in particular, parasol and the Large Off cells, seem to be affected by additional interactions that are not triggered during simple light-intensity flashes. Together, our data show that ganglion cells in the primate retina are sensitive to different combinations of presaccadic and postsaccadic visual stimuli. This contributes to the functional diversity of the output signals of the retina and to asymmetries between On and Off pathways and provides evidence of signal processing beyond what is triggered by isolated steps in light intensity.SIGNIFICANCE STATEMENT Sudden eye movements (saccades) shift our direction of gaze, bringing new images in focus on our retinas. To study how retinal neurons deal with these rapid image transitions, we recorded spiking activity from ganglion cells, the output neurons of the retina, in isolated retinas of marmoset monkeys while shifting a projected image in a saccade-like fashion across the retina. We found that the cells do not just respond to the newly fixated image, but that different types of ganglion cells display different sensitivities to the presaccadic and postsaccadic stimulus patterns. Certain Off cells, for example, are sensitive to changes in the image across transitions, which contributes to differences between On and Off information channels and extends the range of encoded stimulus features.


Asunto(s)
Callithrix , Movimientos Sacádicos , Animales , Retina/fisiología , Movimientos Oculares , Células Ganglionares de la Retina/fisiología , Estimulación Luminosa
12.
J Neurosci ; 43(26): 4808-4820, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37268418

RESUMEN

High-fidelity electronic implants can in principle restore the function of neural circuits by precisely activating neurons via extracellular stimulation. However, direct characterization of the individual electrical sensitivity of a large population of target neurons, to precisely control their activity, can be difficult or impossible. A potential solution is to leverage biophysical principles to infer sensitivity to electrical stimulation from features of spontaneous electrical activity, which can be recorded relatively easily. Here, this approach is developed and its potential value for vision restoration is tested quantitatively using large-scale multielectrode stimulation and recording from retinal ganglion cells (RGCs) of male and female macaque monkeys ex vivo Electrodes recording larger spikes from a given cell exhibited lower stimulation thresholds across cell types, retinas, and eccentricities, with systematic and distinct trends for somas and axons. Thresholds for somatic stimulation increased with distance from the axon initial segment. The dependence of spike probability on injected current was inversely related to threshold, and was substantially steeper for axonal than somatic compartments, which could be identified by their recorded electrical signatures. Dendritic stimulation was largely ineffective for eliciting spikes. These trends were quantitatively reproduced with biophysical simulations. Results from human RGCs were broadly similar. The inference of stimulation sensitivity from recorded electrical features was tested in a data-driven simulation of visual reconstruction, revealing that the approach could significantly improve the function of future high-fidelity retinal implants.SIGNIFICANCE STATEMENT This study demonstrates that individual in situ primate retinal ganglion cells of different types respond to artificially generated, external electrical fields in a systematic manner, in accordance with theoretical predictions, that allows for prediction of electrical stimulus sensitivity from recorded spontaneous activity. It also provides evidence that such an approach could be immensely helpful in the calibration of clinical retinal implants.


Asunto(s)
Retina , Células Ganglionares de la Retina , Animales , Masculino , Femenino , Humanos , Células Ganglionares de la Retina/fisiología , Potenciales de Acción/fisiología , Retina/fisiología , Primates , Estimulación Eléctrica/métodos
13.
Artículo en Inglés | MEDLINE | ID: mdl-39010839

RESUMEN

Gap junctions are channels that allow for direct transmission of electrical signals between cells. However, the ability of one cell to be impacted or controlled by other cells through gap junctions remains unclear. In this study, heterocellular coupling between ON α retinal ganglion cells (RGCs) and displaced amacrine cells (ACs) in the mouse retina was utilized as a model. The impact of the extent of coupling of interconnected ACs on the synchronized firing between coupled ON α RGC-ACs pairs was investigated. It was observed that the synchronized firing between the ON α RGC-ACs pairs was increased by the dopamine 1 receptor antagonist SCH23390, while it was eradicated by the agonist SKF38393. Subsequently, coupled ON α RGC-AC pairs were infected with the channelrhodopsin-2(ChR2) mutation L132C. The spikes of ON α RGCs (without ChR2) could be triggered by ACs (with ChR2) through the gap junction, and vice versa. Furthermore, it was observed that ON α RGCs stimulated with 3-10 Hz currents by whole-cell patch could elicit synchronous spikes in the coupled ACs, and vice versa. The study implies that the synchronized firing between ON α RGC-AC pairs could potentially be affected by the coupling of interconnected ACs, and another cell type could selectively control the firing of one cell type, and information could be forcefully transmitted. The key role of gap junctions in synchronizing firing and driving cells between α RGCs and coupled ACs in the mouse retina is highlighted.

14.
J Transl Med ; 22(1): 447, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741132

RESUMEN

BACKGROUND: Retinal ischemia/reperfusion (RIR) is implicated in various forms of optic neuropathies, yet effective treatments are lacking. RIR leads to the death of retinal ganglion cells (RGCs) and subsequent vision loss, posing detrimental effects on both physical and mental health. Apigenin (API), derived from a wide range of sources, has been reported to exert protective effects against ischemia/reperfusion injuries in various organs, such as the brain, kidney, myocardium, and liver. In this study, we investigated the protective effect of API and its underlying mechanisms on RGC degeneration induced by retinal ischemia/reperfusion (RIR). METHODS: An in vivo model was induced by anterior chamber perfusion following intravitreal injection of API one day prior to the procedure. Meanwhile, an in vitro model was established through 1% oxygen and glucose deprivation. The neuroprotective effects of API were evaluated using H&E staining, spectral-domain optical coherence tomography (SD-OCT), Fluoro-Gold retrograde labeling, and Photopic negative response (PhNR). Furthermore, transmission electron microscopy (TEM) was employed to observe mitochondrial crista morphology and integrity. To elucidate the underlying mechanisms of API, the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, flow cytometry assay, western blot, cell counting kit-8 (CCK-8) assay, lactate dehydrogenase (LDH) assay, JC-1 kit assay, dichlorofluorescein-diacetate (DCFH-DA) assay, as well as TMRE and Mito-tracker staining were conducted. RESULTS: API treatment protected retinal inner plexiform layer (IPL) and ganglion cell complex (GCC), and improved the function of retinal ganglion cells (RGCs). Additionally, API reduced RGC apoptosis and decreased lactate dehydrogenase (LDH) release by upregulating Bcl-2 and Bcl-xL expression, while downregulating Bax and cleaved caspase-3 expression. Furthermore, API increased mitochondrial membrane potential (MMP) and decreased extracellular reactive oxygen species (ROS) production. These effects were achieved by enhancing mitochondrial function, restoring mitochondrial cristae morphology and integrity, and regulating the expression of OPA1, MFN2, and DRP1, thereby regulating mitochondrial dynamics involving fusion and fission. CONCLUSION: API protects RGCs against RIR injury by modulating mitochondrial dynamics, promoting mitochondrial fusion and fission.


Asunto(s)
Apigenina , Dinámicas Mitocondriales , Fármacos Neuroprotectores , Daño por Reperfusión , Células Ganglionares de la Retina , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Apigenina/farmacología , Apigenina/uso terapéutico , Animales , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Dinámicas Mitocondriales/efectos de los fármacos , Masculino , Apoptosis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Modelos Biológicos , Ratones Endogámicos C57BL
15.
Ophthalmology ; 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39111669

RESUMEN

PURPOSE: To examine the relationship between systemic arterial blood pressure (BP) and the rate of change in standard automated perimetry (SAP) in eyes with glaucoma and glaucoma suspects. DESIGN: Prospective cohort study. PARTICIPANTS: 124 eyes (91 glaucoma, 33 suspects) of 64 subjects (mean age 68.4±7.6 years) followed at the Bascom Palmer Eye Institute, Palm Beach Gardens, FL. METHODS: Participants underwent ophthalmic examination, BP measurement, and SAP at 4-month intervals. 24-hour ambulatory blood pressure monitoring (ABPM) was acquired at the baseline visit. Linear mixed models (adjusted for inclusion of both eyes, age, gender, race, intraocular pressure, baseline severity, and central corneal thickness) were used to investigate the effect of BP on the rates of SAP mean deviation (MD) change over time. MAIN OUTCOME MEASURES: Effect of baseline 24-hour and follow-up mean arterial (MAP), systolic (SBP), and diastolic blood pressure (DBP) on change in SAP MD. RESULTS: Eyes had an average of 8.9±1.5 SAP exams over 28.3±6.0 months of follow-up. The median rate of MD change was 0.14dB/year (range -1.21 to 0.96dB/year) with 9 (7%) eyes presenting moderate to fast progression (MD change ≤ -0.50 dB/year). Each 10mmHg lower in 24-hour average MAP and SBP were associated with -0.171 dB/year (P=0.045) and -0.137 dB/year (P=0.023) faster rates of MD loss. Lower mean SBP during follow up was significantly (P = 0.003) associated with MD progression. CONCLUSIONS: Lower baseline 24-hour ABPM measurements, as well as low SBP during follow-up, were significantly associated with faster rates of glaucomatous SAP progression and may be used as a predictor of risk of glaucomatous progression.

16.
Exp Eye Res ; 239: 109754, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38113955

RESUMEN

The purpose of this study was to examine the effect of a blast exposure generated from a shock tube on retinal ganglion cell (RGC) function and structure. Mice were exposed to one of three blast conditions using a shock tube; a single blast wave of 20 PSI, a single blast wave of 30 PSI, or three blast waves of 30 PSI given on three consecutive days with a one-day inter-blast interval. The structure and function of the retina were analyzed using the pattern electroretinogram (PERG), the optomotor reflex (OMR), and optical coherence tomography (OCT). The in vivo parameters were examined at baseline, and then again 1-week, 4-weeks, and 16-weeks following blast exposure. The number of surviving RGCs was quantified at the end of the study. Analysis of mice receiving a 20 PSI injury showed decreased PERG and OMR responses 16-weeks post blast, without evidence of changed retinal thickness or RGC death. Mice subjected to a 30 PSI injury showed decreased PERG responses 4 weeks and 16 weeks after injury, without changes in the retinal thickness or RGC density. Mice subjected to 30 PSI X 3 blast exposures had PERG deficits 1-week and 4-weeks post exposure. There was also significant change in retinal thickness 1-week and 16-weeks post blast exposure. Mice receiving 30 PSI X 3 blast injuries had regional loss of RGCs in the central retina, but not in the mid-peripheral or peripheral retina. Overall, this study has shown that increasing the number of blast exposures and the intensity leads to earlier functional loss of RGCs. We have also shown regional RGC loss only when using the highest blast intensity and number of blast injuries.


Asunto(s)
Traumatismos por Explosión , Células Ganglionares de la Retina , Ratones , Animales , Células Ganglionares de la Retina/metabolismo , Traumatismos por Explosión/metabolismo , Retina , Electrorretinografía , Muerte Celular , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
17.
Exp Eye Res ; 244: 109929, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38750783

RESUMEN

Optic nerve injuries are severely disrupt the structural and functional integrity of the retina, often leading to visual impairment or blindness. Despite the profound impact of these injuries, the molecular mechanisms involved remain poorly understood. In this study, we performed a comprehensive whole-transcriptome analysis of mouse retina samples after optic nerve crush (ONC) to elucidate changes in gene expression and regulatory networks. Transcriptome analysis revealed a variety of molecular alterations, including 256 mRNAs, 530 lncRNAs, and 37 miRNAs, associated with metabolic, inflammatory, signaling, and biosynthetic pathways in the injured retina. The integrated analysis of co-expression and protein-protein interactions identified an active interconnected module comprising 5 co-expressed proteins (Fga, Serpina1a, Hpd, Slc38a4, and Ahsg) associated with the complement and coagulation cascades. Finally, 5 mRNAs (Fga, Serpinala, Hpd, Slc38a4, and Ahsg), 2 miRNAs (miR-671-5p and miR-3057-5p), and 6 lncRNAs (MSTRG. 1830.1, Gm10814, A530013C23Rik, Gm40634, MSTRG.9514.1, A330023F24Rik) were identified by qPCR in the injured retina, and some of them were validated as critical components of a ceRNA network active in 661W and HEK293T cells through dual-luciferase reporter assays. In conclusion, our study provides comprehensive insight into the complex and dynamic biological mechanisms involved in retinal injury responses and highlights promising potential targets to enhance neuroprotection and restore vision.


Asunto(s)
Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Ratones Endogámicos C57BL , Traumatismos del Nervio Óptico , ARN Mensajero , Retina , Animales , Ratones , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/genética , Retina/metabolismo , ARN Mensajero/genética , Modelos Animales de Enfermedad , Transcriptoma , MicroARNs/genética , Regulación de la Expresión Génica/fisiología , Masculino , Humanos , ARN Largo no Codificante/genética
18.
Exp Eye Res ; 239: 109787, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38211683

RESUMEN

Retinal ganglion cell (RGC) death and axonal loss cause irreversible vision loss upon optic nerve (ON) injury. We have independently demonstrated that mesenchymal stem cells (MSCs) and green tea extract (GTE) promote RGC survival and axonal regeneration in rats with ON injury. Here we aimed to evaluate the combined treatment effect of human bone marrow-derived MSCs (hBM-MSCs) and GTE on RGC survival and axonal regeneration after ON injury. Combined treatment of hBM-MSCs and GTE promoted RGC survival and neurite outgrowth/axonal regeneration in ex vivo retinal explant culture and in rats after ON injury. GTE increased Stat3 activation in the retina after combined treatment, and enhanced brain-derived neurotrophic factor secretion from hBM-MSCs. Treatment of 10 µg/mL GTE would not induce hBM-MSC apoptosis, but inhibited their proliferation, migration, and adipogenic and osteogenic differentiation in vitro with reducing matrix metalloproteinase secretions. In summary, this study revealed that GTE can enhance RGC protective effect of hBM-MSCs, suggesting that stem cell priming could be a prospective strategy enhancing the properties of stem cells for ON injury treatment.


Asunto(s)
Células Madre Mesenquimatosas , Traumatismos del Nervio Óptico , Ratas , Humanos , Animales , Traumatismos del Nervio Óptico/terapia , Traumatismos del Nervio Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Osteogénesis , Té/metabolismo , Regeneración Nerviosa/fisiología , Supervivencia Celular/fisiología , Axones/metabolismo
19.
Exp Eye Res ; 246: 110012, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39059735

RESUMEN

Photopic negative response (PhNR), an index of retinal ganglion cell (RGC) function, is impaired in patients with optic pathway gliomas (OPGs). The aim of this longitudinal study was to evaluate whether PhNR deteriorates over time in OPG patients. Fourteen pediatric patients affected by OPG (4 males and 10 females, mean age 12.4 ± 5.7 years, 8 with neurofibromatosis type 1 [NF1]) with ≥12 months of follow-up and ≥2 evaluations, were included in this retrospective study. All patients had received chemotherapy, with or without OPG surgical resection, at least 5 years prior to the study. At baseline, all patients underwent a complete ophthalmological examination. Follow-up included clinical examination and PhNR measurement as well as brain MRI (according to pediatric oncologist indications) every 6 or 12 months. Mean follow-up duration was 16.7 ± 7.5 months (range 12-36 months). Photopic electroretinograms were elicited by 2.0 cd-s/m2 Ganzfeld white flashes presented on a steady 20 cd/m2 white background. The PhNR amplitude was measured as the difference between baseline and the maximal negative amplitude (minimum) of the negative wave, following the photopic b-wave. Compared to baseline, mean PhNR amplitude was significantly decreased at the end of follow-up (p = 0.008). NF1-related OPGs exhibited a decline in PhNR amplitude (p = 0.005) and an increase in PhNR peak-time during the follow-up (p = 0.013), whereas sporadic OPGs showed no significant changes. Tumor size remained stable in all patients on MRI. PhNR amplitude decreased over the observation period, suggesting progressive RGC dysfunction in NF1-related pediatric OPGs, despite stable size on MRI imaging. PhNR could serve as a non-invasive objective tool for assessing longitudinal changes in RGC function in the clinical management of childhood OPG.

20.
Exp Eye Res ; 241: 109834, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38382575

RESUMEN

Myopia and astigmatism are two primary types of refractive errors characterized by inaccurate focusing images on the retina. This study aimed to investigate the response characteristics of Retinal Ganglion Cells (RGCs), represented by alpha (α) RGCs, when exposed to focused, simulated spherically defocused images and astigmatically defocused images projected onto mouse retinas. Negative pressure was applied to stretch the soma of RGC in vitro to simulate myopia using a 7-8 µm diameter glass microelectrode, resulting in a 5% increase in the cell's diameter. A custom-made device was utilized to project spherically (equal to ±10 and ± 20 D) and astigmatically (+6.00 D) defocused images onto the retinas. As a control for a deficient intact retinal circuit, αRGCs of connexin 36 knockout (Cx36 KO) mice were used. The response of αRGCs varied significantly in terms of spikes, excitatory postsynaptic currents (EPSCs) and capacitances under stretching conditions to mimic myopia. Significant differences in the amplitudes of EPSCs were observed in the majority of αRGCs when exposed to focused and spherically defocused images in normal and mechanically simulated myopic retinas. However, this difference was not observed in αRGCs of Cx36 KO mice. αRGCs demonstrated significant differences in response between focused and astigmatically defocused images. Once again, αRGCs of Cx36 KO mice did not display differences. αRGCs have the ability to detect focused, spherically, and astigmatically defocused images and exhibit differential responses ex vivo. Gap junction subunit Cx36 may play a crucial role in transmitting visual signals associated with developing and perceiving refractive errors.


Asunto(s)
Miopía , Células Ganglionares de la Retina , Animales , Ratones , Retina , Uniones Comunicantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA