ABSTRACT
Retinal ganglion cells (RGCs) are the sole output neurons that transmit visual information from the retina to the brain. Diverse insults and pathological states cause degeneration of RGC somas and axons leading to irreversible vision loss. A fundamental question is whether manipulation of a key regulator of RGC survival can protect RGCs from diverse insults and pathological states, and ultimately preserve vision. Here, we report that CaMKII-CREB signaling is compromised after excitotoxic injury to RGC somas or optic nerve injury to RGC axons, and reactivation of this pathway robustly protects RGCs from both injuries. CaMKII activity also promotes RGC survival in the normal retina. Further, reactivation of CaMKII protects RGCs in two glaucoma models where RGCs degenerate from elevated intraocular pressure or genetic deficiency. Last, CaMKII reactivation protects long-distance RGC axon projections in vivo and preserves visual function, from the retina to the visual cortex, and visually guided behavior.
Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cytoprotection , Retinal Ganglion Cells/pathology , Vision, Ocular , Animals , Axons/drug effects , Axons/pathology , Brain/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Dependovirus/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Glaucoma/genetics , Glaucoma/pathology , Mice, Inbred C57BL , Neurotoxins/toxicity , Optic Nerve Injuries/pathology , Signal TransductionABSTRACT
Axon regeneration is an energy-demanding process that requires active mitochondrial transport. In contrast to the central nervous system (CNS), axonal mitochondrial transport in regenerating axons of the peripheral nervous system (PNS) increases within hours and sustains for weeks after injury. Yet, little is known about targeting mitochondria in nervous system repair. Here, we report the induction of sustained axon regeneration, neural activities in the superior colliculus (SC), and visual function recovery after optic nerve crush (ONC) by M1, a small molecule that promotes mitochondrial fusion and transport. We demonstrated that M1 enhanced mitochondrial dynamics in cultured neurons and accelerated in vivo axon regeneration in the PNS. Ex vivo time-lapse imaging and kymograph analysis showed that M1 greatly increased mitochondrial length, axonal mitochondrial motility, and transport velocity in peripheral axons of the sciatic nerves. Following ONC, M1 increased the number of axons regenerating through the optic chiasm into multiple subcortical areas and promoted the recovery of local field potentials in the SC after optogenetic stimulation of retinal ganglion cells, resulting in complete recovery of the pupillary light reflex, and restoration of the response to looming visual stimuli was detected. M1 increased the gene expression of mitochondrial fusion proteins and major axonal transport machinery in both the PNS and CNS neurons without inducing inflammatory responses. The knockdown of two key mitochondrial genes, Opa1 or Mfn2, abolished the growth-promoting effects of M1 after ONC, suggesting that maintaining a highly dynamic mitochondrial population in axons is required for successful CNS axon regeneration.
Subject(s)
Axons , Optic Nerve Injuries , Humans , Axons/metabolism , Mitochondrial Proteins/metabolism , Nerve Crush , Nerve Regeneration/physiology , Optic Nerve/metabolism , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/physiology , Sciatic Nerve/metabolism , Small Molecule LibrariesABSTRACT
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.
Subject(s)
Optic Nerve Injuries , RNA, Circular , Animals , Humans , Disease Models, Animal , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/metabolism , Retina , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , RNA, Circular/genetics , RNA, Circular/metabolismABSTRACT
Optic neuropathies, such as glaucoma, are due to progressive retinal ganglion cells (RGCs) degeneration, result in irreversible vision loss. The promising RGCs replacement therapy for restoring vision are impeded by insufficient RGC-like cells sources. The present work was enriched one new type RGC-like cells using two surface markers CD184 and CD171 from human induced pluripotent stem cells (hiPSCs) by FACS sorting firstly. These new kind cells have well proliferation ability and possessed passage tolerance in vitro 2D or 3D spheroids culture, which kept expressing Pax6, Brn3b and ßIII-Tubulin and so on. The transplanted CD184+CD171+ RGC-like cells could survive and integrate into the normal and optic nerve crush (ONC) mice retina, especially they were more inclined to across the optic nerve head and extend to the damaged optic nerve. These data support the feasible application for cell replacement therapy in RGC degenerative diseases, as well as help to develop new commercial cells sorting reagents and establish good manufacturing practice (GMP) grade RGC-like donor cells for further clinical application.
Subject(s)
Induced Pluripotent Stem Cells , Optic Nerve Injuries , Mice , Animals , Humans , Retina , Retinal Ganglion Cells , Optic Nerve , Organoids , Disease Models, Animal , Nerve CrushABSTRACT
This study demonstrated the functions and molecular mechanisms of the IL-33/ST2 axis in experimental optic neuropathy. C57BL/6J mice were used to establish an optic nerve crush (ONC) model. ONC mice were administered with IL-33 intraperitoneal injection, with PBS vehicle as control. Immunofluorescence, quantitative RT-PCR, and western blot techniques were utilized to assess the expression of the IL-33/ST2 axis. The electroretinography (ERG), optical coherence tomography (OCT), H&E, and luxol fast blue were used to assess the structural and functional changes. Western blot was employed to detect the activation of the mTOR/S6 pathway. The IL-33 expression level in the inner nuclear layer of the retina in ONC mice reached its peak on day 3, accompanied by a significant increase in IL-33 receptor ST2 expression. IL-33 treatment promoted the survival of retinal ganglion cells, restored the thickness of inner retina layer (IRL), alleviated the demyelination of the optic nerve, and recovered the decreased amplitude of b-wave in ONC mice. Furthermore, administration of IL-33 activated the mTOR/S6 signaling pathway in RGCs, which was significantly suppressed in the ONC condition. This study indicated that boosting the IL-33/ST2/mTOR/S6 pathway can protect against structural and functional damage to the retina and optic nerve induced by ONC. As a result, the IL-33/ST2 axis holds potential as a therapeutic option for treating various optic neuropathies.
Subject(s)
Blotting, Western , Disease Models, Animal , Electroretinography , Interleukin-33 , Mice, Inbred C57BL , Nerve Crush , Optic Nerve Injuries , Retinal Ganglion Cells , Signal Transduction , TOR Serine-Threonine Kinases , Tomography, Optical Coherence , Animals , Interleukin-33/metabolism , Mice , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Male , Interleukin-1 Receptor-Like 1 Protein/metabolism , Optic Nerve/metabolism , Optic Nerve/pathology , Retina/metabolismABSTRACT
BACKGROUND: The preservation of retinal ganglion cells (RGCs) and the facilitation of axon regeneration are crucial considerations in the management of various vision-threatening disorders. Therefore, we investigate the efficacy of interleukin-4 (IL-4), a potential therapeutic agent, in promoting neuroprotection and axon regeneration of retinal ganglion cells (RGCs) as identified through whole transcriptome sequencing in an in vitro axon growth model. METHODS: A low concentration of staurosporine (STS) was employed to induce in vitro axon growth. Whole transcriptome sequencing was utilized to identify key target factors involved in the molecular mechanism underlying axon growth. The efficacy of recombinant IL-4 protein on promoting RGC axon growth was validated through in vitro experiments. The protective effect of recombinant IL-4 protein on somas of RGCs was assessed using RBPMS-specific immunofluorescent staining in mouse models with optic nerve crush (ONC) and N-methyl-D-aspartic acid (NMDA) injury. The protective effect on RGC axons was evaluated by anterograde labeling of cholera toxin subunit B (CTB), while the promotion of RGC axon regeneration was assessed through both anterograde labeling of CTB and immunofluorescent staining for growth associated protein-43 (GAP43). RESULTS: Whole-transcriptome sequencing of staurosporine-treated 661 W cells revealed a significant upregulation in intracellular IL-4 transcription levels during the process of axon regeneration. In vitro experiments demonstrated that recombinant IL-4 protein effectively stimulated axon outgrowth. Subsequent immunostaining with RBPMS revealed a significantly higher survival rate of RGCs in the rIL-4 group compared to the vehicle group in both NMDA and ONC injury models. Axonal tracing with CTB confirmed that recombinant IL-4 protein preserved long-distance projection of RGC axons, and there was a notably higher number of surviving axons in the rIL-4 group compared to the vehicle group following NMDA-induced injury. Moreover, intravitreal delivery of recombinant IL-4 protein substantially facilitated RGC axon regeneration after ONC injury. CONCLUSION: The recombinant IL-4 protein exhibits the potential to enhance the survival rate of RGCs, protect RGC axons against NMDA-induced injury, and facilitate axon regeneration following ONC. This study provides an experimental foundation for further investigation and development of therapeutic agents aimed at protecting the optic nerve and promoting axon regeneration.
Subject(s)
Axons , Interleukin-4 , Nerve Regeneration , Retinal Ganglion Cells , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Animals , Interleukin-4/pharmacology , Axons/drug effects , Axons/metabolism , Nerve Regeneration/drug effects , Mice , Mice, Inbred C57BL , Optic Nerve Injuries/pathology , Optic Nerve Injuries/drug therapy , N-Methylaspartate/pharmacology , Staurosporine/pharmacology , Neuroprotective Agents/pharmacology , Recombinant Proteins/pharmacologyABSTRACT
Resident microglia are important to maintain homeostasis in the central nervous system, which includes the retina. The retinal microglia become activated in numerous pathological conditions, but the molecular signatures of these changes are poorly understood. Here, using an approach based on FACS and RNA-seq, we show that microglial gene expression patterns gradually change during RGC degeneration induced by optic nerve injury. Most importantly, we found that the microglial cells strongly expressed Tnf and Il1α, both of which are known to induce neurotoxic reactive astrocytes, and were characterized by Gpr84high -expressing cells in a particular subpopulation. Moreover, ripasudil, a Rho kinase inhibitor, significantly blunted Gpr84 expression and cytokine induction in vitro and in vivo. Finally, GPR84-deficient mice prevented RGC loss in optic nerve-injured retina. These results reveal that Rho kinase-mediated GPR84 alteration strongly contribute to microglial activation and promote neurotoxicity, suggesting that Rho-ROCK and GPR84 signaling may be potential therapeutic targets to prevent the neurotoxic microglial phenotype induced by optic nerve damage, such as occurs in traumatic optic neuropathy and glaucoma.
Subject(s)
Optic Nerve Injuries , Mice , Animals , Microglia/metabolism , Retinal Ganglion Cells , rho-Associated Kinases/metabolism , Neuroglia/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolismABSTRACT
Tauopathy is a neurodegenerative condition associated with oligomeric tau formation through abnormal phosphorylation. We previously showed that tauopathy is involved in death of retinal ganglion cells (RGCs) after optic nerve crush (ONC). It has been proposed that glycogen synthase kinase 3ß (GSK3ß) is involved in the hyperphosphorylation of tau in Alzheimer's disease. To determine the roles of GSK3ß in tauopathy-related death of RGCs, lithium chloride (LiCl), a GSK3ß inhibitor, was injected intravitreally just after ONC. The neuroprotective effects of LiCl were determined by counting Tuj-1-stained RGCs on day 7. Changes of phosphorylated (ser 396) tau in the retina were determined by Simple Western analysis (WES) on day 3. Retinal GSK3ß levels were determined by immunohistochemistry (IHC) and an ELISA. There was a 1.9- and 2.1-fold increase in the levels of phosphorylated tau monomers and dimers on day 3 after ONC. LiCl significantly suppressed the increase in the levels of phosphorylated tau induced by ONC. GSK3ß was mainly present in somas of RGCs, and ELISA showed that retinal levels increased to 2.0-fold on day 7. IHC showed that the GSK3ß expression increased over time and remained in RGCs that were poorly stained by Tuj-1. The GSK3ß and tau expression was colocalized in RGCs. The number of RGCs decreased from 1881 ± 188 (sham control) to 1150 ± 192 cells/mm2 on day 7, and LiCl preserved the levels at 1548 ± 173 cells/mm2. Accordingly, GSK3ß may be a promising target for some optic nerve injuries.
ABSTRACT
Electroretinogram (ERG) is the most common clinical and basic visual electrodiagnostic test, which has long been used to evaluate the retinal function through photic stimulation. Despite its wide application, there are still some pitfalls often neglected in ERG recording, such as the recording time point, active electrode location, and the animal strain. In this study, we systematically analyzed and compared the effects of multiple factors on ERG, which would provide an important reference for ERG detection by other investigators. ERG was recorded using the Celeris D430 rodent ERG testing system. The amplitudes and latencies of a wave, b wave and oscillatory potentials (OPs) recorded from different electrode locations (subdermal and invasive), different times of day (day time 8:00 to 13:00 and night time 18:00 to 23:00), bilateral eyes (left and right), and different mouse strains (C57 and CD1) were analyzed and compared. Our results revealed that ERG was affected by active electrode locations and difference between day and night, while OPs seemed not to be influenced. There was no significant difference in the amplitudes or latencies of ERG and OPs between left and right eyes, irrespective of measurements at day or night, or which method was used. Compared to C57 mice, both ERG and OP responses were significantly decreased in Brn3bAP/AP mice, a model for retinal ganglion cell (RGC) loss. In addition, there were some non-negligible differences in visual responses between C57 and CD1 mouse strains. Our results suggest that the invasive procedure is a reliable method for evaluating the visual function including VEP, ERG and OP responses in mice. Moreover, these comparative analyses provide valuable references for future studies of mammalian visual electrophysiology.
Subject(s)
Electroretinography , Retina , Mice , Animals , Electroretinography/methods , Retinal Ganglion Cells/physiology , MammalsABSTRACT
The main purpose of this study is to analyze the effects of unilateral optic nerve crush in the gene expression of pro- and anti-inflammatory mediators, and gliosis markers in injured and contralateral retinas. Retinas from intact, unilaterally optic nerve injured or sham-operated C57BL/6J mice were analyzed 1, 3, 9 and 30 days after the surgery (n = 5/group and time point) and the relative expression of TGF-ß1, IL-1ß, TNF-α, Iba1, AQP4, GFAP, MHCII, and TSPO was analyzed in injured and contralateral using qPCR. The results indicated that compared with intact retinas, sham-operated animals showed an early (day 1) upregulation of IL-1ß, TNF-α and TSPO and a late (day 30) upregulation of TNF-α. In sham-contralateral retinas, TNF-α and TSPO mRNA expression were upregulated and day 30 while GFAP, Iba1, AQP4 and MHCII downregulated at day 9. Compared with sham-operated animals, in retinas affected by optic nerve crush GFAP and TSPO upregulated at day 1 and TNF-α, Iba1, AQP4 and MHCII at day 3. In the crushed-contralateral retinas, TGF-ß1, TNF-α, Iba1 and MHCII were upregulated at day 1. TSPO was upregulated up to day 30 whereas TGF-ß1 and Iba1 downregulated after day 9. In conclusion, both sham surgery and optic nerve crush changed the profile of inflammatory and gliosis markers in the injured and contralateral retinas, changes that were more pronounced for optic nerve crush when compared to sham.
Subject(s)
Optic Nerve Injuries , Transforming Growth Factor beta1 , Mice , Animals , Transforming Growth Factor beta1/pharmacology , Retinal Ganglion Cells/metabolism , Gliosis/metabolism , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Neuroinflammatory Diseases , Tumor Necrosis Factor-alpha/metabolism , Mice, Inbred C57BL , Retina/metabolism , Optic Nerve/metabolism , Nerve Crush/methodsABSTRACT
Traumatic optic neuropathy (TON) is a severe condition characterized by retinal ganglion cell (RGC) death, often leading to irreversible vision loss, and the death of RGCs is closely associated with oxidative stress. Unfortunately, effective treatment options for TON are lacking. To address this, catalase (CAT) is encapsulated in a tannic acid (TA)/poly(ethylenimine)-crosslinked hollow nanoreactor (CAT@PTP), which exhibited enhanced anchoring in the retina due to TA-collagen adhesion. The antioxidative activity of both CAT and TA synergistically eliminated reactive oxygen species (ROS) to save RGCs in the retina, thereby treating TON. In vitro experiments demonstrated that the nanoreactors preserve the enzymatic activity of CAT and exhibit high adhesion to type I collagen. The combination of CAT and TA-based nanoreactors enhanced ROS elimination while maintaining high biocompatibility. In an optic nerve crush rat model, CAT@PTP is effectively anchored to the retina via TA-collagen adhesion after a single vitreous injection, and RGCs are significantly preserved without adverse events. CAT@PTP exhibited a protective effect on retinal function. Given the abundance of collagen that exists in ocular tissues, these findings may contribute to the further application of this multifunctional nanoreactor in ocular diseases to improve therapeutic efficacy and reduce adverse effects.
Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Rats , Animals , Retinal Ganglion Cells/metabolism , Collagen Type I/metabolism , Reactive Oxygen Species/metabolism , Optic Nerve/metabolism , Optic Nerve Injuries/metabolism , Nanotechnology , Cell Survival , Disease Models, AnimalABSTRACT
PURPOSE: The uniform field electroretinogram (UF-ERG) has been suggested as an alternative to the pattern electroretinogram (PERG) for non-invasive assessment of retinal ganglion cell (RGC) function in primates. We evaluated the validity of the UF-ERG to assess mouse RGC activity in vivo. METHODS: Unilateral optic nerve crush (ONC) was performed on adult C57BL/6J mice. Contralateral eyes with uncrushed optic nerves and eyes from surgically naive mice served as experimental controls. Electrophysiological visual assessment was performed at 12 weeks post-ONC. Flash-mediated visual-evoked cortical potentials (VEPs) were measured to confirm the robustness of the ONC procedure. Full-field flash ERGs were used to interrogate photoreceptor and retinal bipolar cell function. RGC function was assessed with pattern ERGs. Summed onset and offset UF-ERG responses to alternating dark and light uniform field flash stimuli of different intensities and wavelengths were recorded from ONC and control eyes, and relative differences were compared to the PERG results. Following electrophysiological analysis, RGC loss was monitored by immunohistochemical staining of the RGC marker protein, RBPMS, in post-mortem retinal tissues. RESULTS: ONC dramatically impacts RGC integrity and optic nerve function, demonstrated by reduced RGC counts and near complete elimination of VEPs. ONC did not affect scotopic ERG a-wave and b-wave amplitudes, while PERG amplitudes of eyes subjected to ONC were reduced by approximately 50% compared to controls. Summation of ON and OFF UF-ERG responses did not reveal statistically significant differences between ONC and control eyes, regardless of visual stimulus. CONCLUSIONS: PERG responses are markedly impaired upon ONC, while UF-ERG responses are not significantly affected by surgical trauma to RGC axons in mice. The more closely related pattern and uniform field ERGs recorded in primates suggests species-specific differences in RGC features or subpopulations corresponding to PERG and UF-ERG response generators, limiting the utility of the UF-ERG for mouse RGC functional analysis.
Subject(s)
Electroretinography , Retinal Ganglion Cells , Mice , Animals , Retinal Ganglion Cells/physiology , Electroretinography/methods , Mice, Inbred C57BL , Retina , Optic Nerve , Disease Models, AnimalABSTRACT
PURPOSE: We aim to investigate the effect of Park7 on mice RGC survival and function following optic nerve crush (ONC), and to explore its potential mechanism. METHODS: Wild-type male C57BL/6J mice were subjected to optic nerve crush. Six weeks before ONC, mice received rAAV-shRNA (Park7)-EGFP or rAAV-EGFP intravitreally. Western blotting was used to detect Park7 levels. RGC survival was measured using immunofluorescence. Retinal cell apoptosis was detected using terminal deoxynucleotidyl transferase nick-end-labelling. An electroretinogram (ERG) and the optomotor response (OMR) were used to assess RGC function. Kelch-like ECH-associated protein 1 (Keap1), nuclear factor erythroid 2-related factor (Nrf2), and heme oxygenase 1 (HO-1) levels were assessed using western blotting. RESULTS: ONC injury increased the relative expression of Park7 significantly and decreased RGC survival, the amplitude of the photopic negative response (PhNR), and OMR. Intravitreal injection of rAAV-shRNA(Park7)-EGFP downregulated Park7 expression and was clearly demonstrated by the green fluorescence protein in many retinal layers. Moreover, Park7 downregulation aggravated the decrease in RGC survival and amplitude of PhNR as well as the visual acuity after ONC. However, inhibition of Park7 significantly increased Keap1 levels, decreased the total and nuclear Nrf2 levels, and reduced HO-1 levels. CONCLUSIONS: Park7 downregulation enhanced RGC injury and decreased retinal electrophysiological response and OMR after ONC in mice via the Keap1-Nrf2-HO-1 signaling pathway. Park7 may have neuroprotective effects and could represent a novel way to treat optic neuropathy.
Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Animals , Male , Mice , Disease Models, Animal , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Inbred C57BL , Nerve Crush , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/pharmacology , Optic Nerve/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Signal TransductionABSTRACT
Diabetic retinopathy (DR), the most common microvascular compilation of diabetes, is the leading cause of vision loss and blindness worldwide. Recent studies indicate that retinal neuron impairment occurs before any noticeable vascular changes in DR, and retinal ganglion cell (RGC) degeneration is one of the earliest signs. Axons of RGCs have little capacity to regenerate after injury, clinically leading the visual functional defects to become irreversible. In the past two decades, tremendous progress has been achieved to enable RGC axon regeneration in animal models of optic nerve injury, which holds promise for neural repair and visual restoration in DR. This review summarizes these advances and discusses the potential and challenges for developing optic nerve regeneration strategies treating DR.
Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Optic Nerve Injuries , Animals , Axons/physiology , Nerve Regeneration/physiology , Blindness , Disease Models, AnimalABSTRACT
It is extremely difficult to achieve functional recovery after axonal injury in the adult central nervous system. The activation of G-protein coupled receptor 110 (GPR110, ADGRF1) has been shown to stimulate neurite extension in developing neurons and after axonal injury in adult mice. Here, we demonstrate that GPR110 activation partially restores visual function impaired by optic nerve injury in adult mice. Intravitreal injection of GPR110 ligands, synaptamide and its stable analogue dimethylsynaptamide (A8) after optic nerve crush significantly reduced axonal degeneration and improved axonal integrity and visual function in wild-type but not gpr110 knockout mice. The retina obtained from the injured mice treated with GPR110 ligands also showed a significant reduction in the crush-induced loss of retinal ganglion cells. Our data suggest that targeting GPR110 may be a viable strategy for functional recovery after optic nerve injury.
Subject(s)
Optic Nerve Injuries , Animals , Mice , Axons , Ligands , Mice, Knockout , Nerve Crush , Nerve Regeneration/physiology , Receptors, G-Protein-Coupled/genetics , Retina , Retinal Ganglion Cells/physiologyABSTRACT
Glaucomatous optic neuropathy (GON), a major cause of blindness, is characterized by the loss of retinal ganglion cells (RGCs) and the degeneration of their axons. Mitochondria are deeply involved in maintaining the health of RGCs and their axons. Therefore, lots of attempts have been made to develop diagnostic tools and therapies targeting mitochondria. Recently, we reported that mitochondria are uniformly distributed in the unmyelinated axons of RGCs, possibly owing to the ATP gradient. Thus, using transgenic mice expressing yellow fluorescent protein targeting mitochondria exclusively in RGCs within the retina, we assessed the alteration of mitochondrial distributions induced by optic nerve crush (ONC) via in vitro flat-mount retinal sections and in vivo fundus images captured with a confocal scanning ophthalmoscope. We observed that the mitochondrial distribution in the unmyelinated axons of survived RGCs after ONC remained uniform, although their density increased. Furthermore, via in vitro analysis, we discovered that the mitochondrial size is attenuated following ONC. These results suggest that ONC induces mitochondrial fission without disrupting the uniform mitochondrial distribution, possibly preventing axonal degeneration and apoptosis. The in vivo visualization system of axonal mitochondria in RGCs may be applicable in the detection of the progression of GON in animal studies and potentially in humans.
Subject(s)
Glaucoma , Optic Nerve Diseases , Optic Nerve Injuries , Mice , Humans , Animals , Retinal Ganglion Cells/metabolism , Optic Nerve Injuries/metabolism , Mitochondrial Dynamics , Glaucoma/metabolism , Optic Nerve Diseases/metabolism , Axons/metabolism , Mice, Transgenic , Disease Models, Animal , Mitochondria/metabolismABSTRACT
PURPOSE: To explore the pathological changes in optic nerve injury models under varying forces. METHODS: The rats were classified into 4 groups: sham operation (SH), 0.1, 0.3, and 0.5 N. Modeling was performed using the lateral optic nerve pulling method. Seven days after modeling, Brn3a immunofluorescence was used to detect retinal ganglion cell (RGC) number, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining was used to detect RGC apoptosis, and flash visual evoked potential (FVEP) was used to detect the optic nerve function on days 1, 3, and 7 after modeling. In addition, LC3 II and P62 expression levels in retinal tissues were detected by western blotting to observe the changes in autophagy levels. RESULTS: RGC number decreased 7 d after modeling, and it showed a downward trend with increasing damaging force. The number of apoptotic RGCs in ganglion cell layer in the 0.3 and 0.5 N groups was increased and was higher than that in the 0.1 N group. The difference in FVEP of rats in each group was mainly reflected in the P2 peak latency. LC3 II and P62 expression levels in retinal tissue of 0.3 and 0.5 N groups were higher than those of the SH and 0.1 groups; however, the difference between the 0.1 N and SH groups was not statistically significant. CONCLUSION: Precisely controlling the force of the optic nerve clamping injury model is necessary because different forces acting on the optic nerve will lead to differences in the loss of optic neurons, the conduction function of the optic nerve, and autophagy level in retinal tissues.
Subject(s)
Optic Nerve Injuries , Rats , Animals , Evoked Potentials, Visual , Retina/pathology , Retinal Ganglion Cells/pathology , Optic Nerve/pathology , Disease Models, AnimalABSTRACT
Analysis of retina cell type-specific epigenetic and transcriptomic signatures is crucial to understanding the pathophysiology of retinal degenerations such as age-related macular degeneration (AMD) and delineating cell autonomous and cell-non-autonomous mechanisms. We have discovered that Aldh1l1 is specifically expressed in the major macroglia of the retina, Müller glia, and, unlike the brain, is not expressed in retinal astrocytes. This allows use of Aldh1l1 cre drivers and Nuclear Tagging and Translating Ribosome Affinity Purification (NuTRAP) constructs for temporally controlled labeling and paired analysis of Müller glia epigenomes and translatomes. As validated through a variety of approaches, the Aldh1l1cre/ERT2-NuTRAP model provides Müller glia specific translatomic and epigenomic profiles without the need to isolate whole cells. Application of this approach to models of acute injury (optic nerve crush) and chronic stress (aging) uncovered few common Müller glia-specific transcriptome changes in inflammatory pathways, and mostly differential signatures for each stimulus. The expression of members of the IL-6 and integrin-linked kinase signaling pathways was enhanced in Müller glia in response to optic nerve crush but not aging. Unique changes in neuroinflammation and fibrosis signaling pathways were observed in response to aging but not with optic nerve crush. The Aldh1l1cre/ERT2-NuTRAP model allows focused molecular analyses of a single, minority cell type within the retina, providing more substantial effect sizes than whole tissue analyses. The NuTRAP model, nucleic acid isolation, and validation approaches presented here can be applied to any retina cell type for which a cell type-specific cre is available.
Subject(s)
Retina , Retinal Degeneration , Humans , Retina/metabolism , Neuroglia/metabolism , Retinal Degeneration/metabolism , Nerve Crush , Optic NerveABSTRACT
The pathological basis of optic nerve crush (ONC) is the apoptosis of retinal ganglion cells (RGCs), which leads to an irreversible impairment of visual function. When stimulated by external stimuli, microglia polarize into different types and play different roles in repairing retinal injury. In this study, gadolinium chloride (GdCl3) could inhibit the excessive proliferation and activation of microglia in the retina after ONC and significantly inhibited the morphological changes of microglia in the ganglion cell layer (GCL) and inner plexiform layer (IPL). In the early stage of optic nerve injury, blood-derived immune cells did not play an essential role in retinal repair. In addition, transcriptome analysis showed that GdCl3 inhibited the expression of microglia proliferation-related factors and regulated signaling pathways related to skeletonization and inflammation. After GdCl3 treatment, M1 markers were significantly down-regulated, while M2 markers were increased. In conclusion, this study demonstrated that GdCl3 could regulate the distribution and morphological change of the retinal microglia and protect the ganglion cells by eliminating M1 microglia selectively, which provided a theoretical basis for further localizing different types of microglia in retina related diseases.
Subject(s)
Microglia , Optic Nerve Injuries , Gadolinium , Humans , Microglia/metabolism , Nerve Crush , Optic Nerve/metabolism , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/metabolismABSTRACT
Visual evoked potential (VEP) is commonly used to evaluate visual acuity in both clinical and basic studies. Subdermal needle electrodes or skull pre-implanted screw electrodes are usually used to record VEP in rodents. However, the VEP amplitudes recorded by the former are small while the latter may damage the brain. In this study, we established a new invasive procedure for VEP recording, and made a series of comparisons of VEP parameters recorded from different electrode locations, different times of day (day and night) and bilateral eyes, to evaluate the influence of these factors on VEP in mice. Our data reveal that our invasive method is reliable and can record VEP with good waveforms and large amplitudes. The comparison data show that VEP is greatly influenced by active electrode locations and difference between day and night. In C57 or CD1 ONC (optic nerve crush) models and Brn3bAP/AP mice, which are featured by loss of retinal ganglion cells (RGCs), amplitudes of VEP N1 and P1 waves are drastically reduced. The newly established VEP procedure is very reliable and stable, and is particularly useful for detecting losses of RGC quantities, functions or connections to the brain. Our analyses of various recording conditions also provide useful references for future studies.