Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38617277

ABSTRACT

Optineurin (OPTN) mutations are linked to amyotrophic lateral sclerosis (ALS) and normal tension glaucoma (NTG), but a relevant animal model is lacking, and the molecular mechanisms underlying neurodegeneration are unknown. We found that OPTN C-terminus truncation (OPTN∆C) causes late-onset neurodegeneration of retinal ganglion cells (RGCs), optic nerve (ON), and spinal cord motor neurons, preceded by a striking decrease of axonal mitochondria. Surprisingly, we discover that OPTN directly interacts with both microtubules and the mitochondrial transport complex TRAK1/KIF5B, stabilizing them for proper anterograde axonal mitochondrial transport, in a C-terminus dependent manner. Encouragingly, overexpressing OPTN/TRAK1/KIF5B reverses not only OPTN truncation-induced, but also ocular hypertension-induced neurodegeneration, and promotes striking ON regeneration. Therefore, in addition to generating new animal models for NTG and ALS, our results establish OPTN as a novel facilitator of the microtubule-dependent mitochondrial transport necessary for adequate axonal mitochondria delivery, and its loss as the likely molecular mechanism of neurodegeneration.

2.
Cell Rep ; 42(6): 112596, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37269288

ABSTRACT

Neural progenitor cells lengthen their cell cycle to prime themselves for differentiation as development proceeds. It is currently not clear how they counter this lengthening and avoid being halted in the cell cycle. We show that N6-methyladenosine (m6A) methylation of cell-cycle-related mRNAs ensures the proper cell-cycle progression of late-born retinal progenitor cells (RPCs), which are born toward the end of retinogenesis and have long cell-cycle length. Conditional deletion of Mettl14, which is required for depositing m6A, led to delayed cell-cycle exit of late-born RPCs but has no effect on retinal development prior to birth. m6A sequencing and single-cell transcriptomics revealed that mRNAs involved in elongating the cell cycle were highly enriched for m6A, which could target them for degradation and guarantee proper cell-cycle progression. In addition, we identified Zfp292 as a target of m6A and potent inhibitor of RPC cell-cycle progression.


Subject(s)
Neural Stem Cells , Retina , Retina/metabolism , Cell Differentiation , Cell Division , Organogenesis
3.
Invest Ophthalmol Vis Sci ; 64(3): 4, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36862119

ABSTRACT

Purpose: Axon transport of organelles and neurotrophic factors is necessary for maintaining cellular function and survival of retinal ganglion cells (RGCs). However, it is not clear how trafficking of mitochondria, essential for RGC growth and maturation, changes during RGC development. The purpose of this study was to understand the dynamics and regulation of mitochondrial transport during RGC maturation using acutely purified RGCs as a model system. Methods: Primary RGCs were immunopanned from rats of either sex during three stages of development. MitoTracker dye and live-cell imaging were used to quantify mitochondrial motility. Analysis of single-cell RNA sequencing was used to identify Kinesin family member 5A (Kif5a) as a relevant motor candidate for mitochondrial transport. Kif5a expression was manipulated with either short hairpin RNA (shRNA) or exogenous expression adeno-associated virus viral vectors. Results: Anterograde and retrograde mitochondrial trafficking and motility decreased through RGC development. Similarly, the expression of Kif5a, a motor protein that transports mitochondria, also decreased during development. Kif5a knockdown decreased anterograde mitochondrial transport, while Kif5a expression increased general mitochondrial motility and anterograde mitochondrial transport. Conclusions: Our results suggested that Kif5a directly regulates mitochondrial axonal transport in developing RGCs. Future work exploring the role of Kif5a in vivo in RGCs is indicated.


Subject(s)
Mitochondria , Retinal Ganglion Cells , Animals , Rats , Axonal Transport , Kinesins/genetics , Models, Biological , RNA, Small Interfering/genetics
4.
Sci Rep ; 12(1): 17446, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36261683

ABSTRACT

Adult central nervous system (CNS) axons fail to regenerate after injury, and master regulators of the regenerative program remain to be identified. We analyzed the transcriptomes of retinal ganglion cells (RGCs) at 1 and 5 days after optic nerve injury with and without a cocktail of strongly pro-regenerative factors to discover genes that regulate survival and regeneration. We used advanced bioinformatic analysis to identify the top transcriptional regulators of upstream genes and cross-referenced these with the regulators upstream of genes differentially expressed between embryonic RGCs that exhibit robust axon growth vs. postnatal RGCs where this potential has been lost. We established the transcriptional activator Elk-1 as the top regulator of RGC gene expression associated with axon outgrowth in both models. We demonstrate that Elk-1 is necessary and sufficient to promote RGC neuroprotection and regeneration in vivo, and is enhanced by manipulating specific phosphorylation sites. Finally, we co-manipulated Elk-1, PTEN, and REST, another transcription factor discovered in our analysis, and found Elk-1 to be downstream of PTEN and inhibited by REST in the survival and axon regenerative pathway in RGCs. These results uncover the basic mechanisms of regulation of survival and axon growth and reveal a novel, potent therapeutic strategy to promote neuroprotection and regeneration in the adult CNS.


Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Humans , Retinal Ganglion Cells/metabolism , Axons/metabolism , Nerve Regeneration/physiology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Transcription Factors/metabolism
5.
J Neurosci ; 42(19): 4042-4052, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35396330

ABSTRACT

Retinal ganglion cells (RGCs) die after optic nerve trauma or in degenerative disease. However, acute changes in protein expression that may regulate RGC response to injury are not fully understood, and detailed methods to quantify new protein synthesis have not been tested. Here, we develop and apply a new in vivo quantitative measure of newly synthesized proteins to examine changes occurring in the retina after optic nerve injury. Azidohomoalanine, a noncanonical amino acid, was injected intravitreally into the eyes of rodents of either sex with or without optic nerve injury. Isotope variants of biotin-alkyne were used for quantitative BONCAT (QBONCAT) mass spectrometry, allowing identification of protein synthesis and transport rate changes in more than 1000 proteins at 1 or 5 d after optic nerve injury. In vitro screening showed several newly synthesized proteins regulate axon outgrowth in primary neurons in vitro This novel approach to targeted quantification of newly synthesized proteins after injury uncovers a dynamic translational response within broader proteostasis regulation and enhances our understanding of the cellular response to injury.SIGNIFICANCE STATEMENT Optic nerve injury results in death and degeneration of retinal ganglion cells and their axons. The specific cellular response to injury, including changes in new protein synthesis, is obscured by existing proteins and protein degradation. In this study, we introduce QBONCAT to isolate and quantify acute protein synthesis and subsequent transport between cellular compartments. We identify novel candidate protein effectors of the regenerative response and uncover their regulation of axon growth in vitro, validating the utility of QBONCAT for the discovery of novel regulatory and therapeutic candidates after optic nerve injury.


Subject(s)
Optic Nerve Injuries , Axons/metabolism , Humans , Nerve Regeneration/physiology , Optic Nerve Injuries/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism
6.
Elife ; 112022 03 08.
Article in English | MEDLINE | ID: mdl-35259089

ABSTRACT

Many neurons in the adult central nervous system, including retinal ganglion cells (RGCs), degenerate and die after injury. Early axon protein and organelle trafficking failure is a key component in many neurodegenerative disorders yet changes to axoplasmic transport in disease models have not been quantified. We analyzed early changes in the protein 'transportome' from RGC somas to their axons after optic nerve injury and identified transport failure of an anterograde motor protein Kif5a early in RGC degeneration. We demonstrated that manipulating Kif5a expression affects anterograde mitochondrial trafficking in RGCs and characterized axon transport in Kif5a knockout mice to identify proteins whose axon localization was Kif5a-dependent. Finally, we found that knockout of Kif5a in RGCs resulted in progressive RGC degeneration in the absence of injury. Together with expression data localizing Kif5a to human RGCs, these data identify Kif5a transport failure as a cause of RGC neurodegeneration and point to a mechanism for future therapeutics.


Subject(s)
Optic Nerve Injuries , Animals , Axonal Transport , Axons/metabolism , Kinesins/genetics , Mice , Mice, Inbred C57BL , Nerve Regeneration , Retinal Ganglion Cells/metabolism
7.
Cell Rep ; 38(4): 110287, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35081342

ABSTRACT

Intercellular transfer of toxic proteins between neurons is thought to contribute to neurodegenerative disease, but whether direct interneuronal protein transfer occurs in the healthy brain is not clear. To assess the prevalence and identity of transferred proteins and the cellular specificity of transfer, we biotinylated retinal ganglion cell proteins in vivo and examined biotinylated proteins transported through the rodent visual circuit using microscopy, biochemistry, and mass spectrometry. Electron microscopy demonstrated preferential transfer of biotinylated proteins from retinogeniculate inputs to excitatory lateral geniculate nucleus (LGN) neurons compared with GABAergic neurons. An unbiased mass spectrometry-based screen identified ∼200 transneuronally transported proteins (TNTPs) isolated from the visual cortex. The majority of TNTPs are present in neuronal exosomes, and virally expressed TNTPs, including tau and ß-synuclein, were detected in isolated exosomes and postsynaptic neurons. Our data demonstrate transfer of diverse endogenous proteins between neurons in the healthy intact brain and suggest that TNTP transport may be mediated by exosomes.


Subject(s)
Cell Communication/physiology , Exosomes/metabolism , Neurons/metabolism , Visual Cortex/metabolism , Animals , Neuroanatomical Tract-Tracing Techniques , Proteomics , Rats , Rats, Wistar , Visual Pathways/metabolism , Xenopus
8.
Am J Ophthalmol ; 222: 76-81, 2021 02.
Article in English | MEDLINE | ID: mdl-32980331

ABSTRACT

PURPOSE: The global COVID-19 pandemic has resulted in a renewed focus on the importance of personal protective equipment (PPE) and other interventions to decrease spread of infectious diseases. Although several ophthalmology organizations have released guidance on appropriate PPE for surgical procedures and ophthalmology clinics, there is limited experimental evidence that demonstrates the efficacy of various interventions that have been suggested. In this study, we evaluated high-risk aspects of the slit-lamp exam and the effect of various PPE interventions, specifically the use of a surgical mask and a slit-lamp shield. DESIGN: Experimental simulation study. METHODS: This was a single-center study in a patient simulation population. This study examined the presence of particles in the air near or on a slit-lamp, a simulated slit-lamp examiner, or a simulated patient using a fluorescent surrogate of respiratory droplets. RESULTS: Simulated coughing without a mask or slit-lamp shield resulted in widespread dispersion of fluorescent droplets during the model slit-lamp examination. Coughing with a mask resulted in the most significant decrease in droplets; however, particles still escaped from the top of the mask. Coughing with the slit-lamp shield alone blocked most of forward particle dispersion; however, significant distributions of respiratory droplets were found on the slit-lamp joystick and table. Coughing with both a mask and slit-lamp shield resulted in the least dispersion to the simulated examiner and the simulated patient. Scanning electron microscopy demonstrated particle sizes of 3-100 µm. CONCLUSIONS: Masking had the greatest effect in limiting spread of respiratory droplets, whereas slit-lamp shields and gloves also contributed to limiting exposure to droplets from SARS-CoV-2 during slit-lamp examination.


Subject(s)
COVID-19/transmission , Disease Transmission, Infectious/prevention & control , Patient Simulation , Personal Protective Equipment , Printing, Three-Dimensional , SARS-CoV-2 , Slit Lamp Microscopy/methods , COVID-19/epidemiology , Humans , Pandemics
9.
PLoS One ; 15(12): e0242884, 2020.
Article in English | MEDLINE | ID: mdl-33315889

ABSTRACT

Loss of retinal ganglion cells (RGCs) in optic neuropathies results in permanent partial or complete blindness. Myocyte enhancer factor 2 (MEF2) transcription factors have been shown to play a pivotal role in neuronal systems, and in particular MEF2A knockout was shown to enhance RGC survival after optic nerve crush injury. Here we expanded these prior data to study bi-allelic, tri-allelic and heterozygous allele deletion. We observed that deletion of all MEF2A, MEF2C, and MEF2D alleles had no effect on RGC survival during development. Our extended experiments suggest that the majority of the neuroprotective effect was conferred by complete deletion of MEF2A but that MEF2D knockout, although not sufficient to increase RGC survival on its own, increased the positive effect of MEF2A knockout. Conversely, MEF2A over-expression in wildtype mice worsened RGC survival after optic nerve crush. Interestingly, MEF2 transcription factors are regulated by post-translational modification, including by calcineurin-catalyzed dephosphorylation of MEF2A Ser-408 known to increase MEF2A-dependent transactivation in neurons. However, neither phospho-mimetic nor phospho-ablative mutation of MEF2A Ser-408 affected the ability of MEF2A to promote RGC death in vivo after optic nerve injury. Together these findings demonstrate that MEF2 gene expression opposes RGC survival following axon injury in a complex hierarchy, and further support the hypothesis that loss of or interference with MEF2A expression might be beneficial for RGC neuroprotection in diseases such as glaucoma and other optic neuropathies.


Subject(s)
MEF2 Transcription Factors/metabolism , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/pathology , Alleles , Animals , Cell Count , Humans , MEF2 Transcription Factors/deficiency , MEF2 Transcription Factors/genetics , Mice , Optic Nerve Injuries/genetics , Point Mutation , Signal Transduction
10.
J Clin Med ; 9(9)2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32957639

ABSTRACT

Various breathing and cough simulators have been used to model respiratory droplet dispersion and viral droplets, in particular for SARS-CoV-2 modeling. However, limited data are available comparing these cough simulations to physiological breathing and coughing. In this study, three different cough simulators (Teleflex Mucosal Atomization Device Nasal (MAD Nasal), a spray gun, and GloGermTM MIST) that have been used in the literature were studied to assess their physiologic relevance. Droplet size, velocity, dispersion, and force generated by the simulators were measured. Droplet size was measured with scanning electron microscopy (SEM). Slow-motion videography was used to 3D reconstruct and measure the velocity of each simulated cough. A force-sensitive resistor was used to measure the force of each simulated cough. The average size of droplets from each cough simulator was 176 to 220 µm. MAD Nasal, the spray gun, and GloGermTM MIST traveled 0.38 m, 0.89 m, and 1.62 m respectively. The average velocities for the MAD Nasal, spray gun, and GloGermTM MIST were 1.57 m/s, 2.60 m/s, and 9.27 m/s respectively, and all yielded a force of <0.5 Newtons. GloGermTM MIST and the spray gun most closely resemble physiological coughs and breathing respectively. In conclusion, none of the simulators tested accurately modeled all physiologic characteristics (droplet size, 3-D dispersion velocity, and force) of a cough, while there were various strengths and weaknesses of each method. One should take this into account when performing simulations with these devices.

11.
Cell Rep ; 28(7): 1935-1947.e5, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412257

ABSTRACT

The brain processes information and generates cognitive and motor outputs through functions of spatially organized proteins in different types of neurons. More complete knowledge of proteins and their distributions within neuronal compartments in intact circuits would help in the understanding of brain function. We used unbiased in vivo protein labeling with intravitreal NHS-biotin for discovery and analysis of endogenous axonally transported proteins in the visual system using tandem mass spectrometric proteomics, biochemistry, and both light and electron microscopy. Purification and proteomic analysis of biotinylated peptides identified ∼1,000 proteins transported from retinal ganglion cells into the optic nerve and ∼575 biotinylated proteins recovered from presynaptic compartments of lateral geniculate nucleus and superior colliculus. Approximately 360 biotinylated proteins were differentially detected in the two retinal targets. This study characterizes axonally transported proteins in the healthy adult visual system by analyzing proteomes from multiple compartments of retinal ganglion cell projections in the intact brain.


Subject(s)
Axonal Transport , Optic Nerve/metabolism , Presynaptic Terminals/metabolism , Proteome/analysis , Proteome/metabolism , Retinal Ganglion Cells/metabolism , Visual Pathways/metabolism , Animals , Male , Optic Nerve/cytology , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Visual Pathways/cytology
12.
J Neurosci ; 39(28): 5466-5480, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31097623

ABSTRACT

cAMP signaling is known to be critical in neuronal survival and axon growth. Increasingly the subcellular compartmentation of cAMP signaling has been appreciated, but outside of dendritic synaptic regulation, few cAMP compartments have been defined in terms of molecular composition or function in neurons. Specificity in cAMP signaling is conferred in large part by A-kinase anchoring proteins (AKAPs) that localize protein kinase A and other signaling enzymes to discrete intracellular compartments. We now reveal that cAMP signaling within a perinuclear neuronal compartment organized by the large multivalent scaffold protein mAKAPα promotes neuronal survival and axon growth. mAKAPα signalosome function is explored using new molecular tools designed to specifically alter local cAMP levels as studied by live-cell FRET imaging. In addition, enhancement of mAKAPα-associated cAMP signaling by isoform-specific displacement of bound phosphodiesterase is demonstrated to increase retinal ganglion cell survival in vivo in mice of both sexes following optic nerve crush injury. These findings define a novel neuronal compartment that confers cAMP regulation of neuroprotection and axon growth and that may be therapeutically targeted in disease.SIGNIFICANCE STATEMENT cAMP is a second messenger responsible for the regulation of diverse cellular processes including neuronal neurite extension and survival following injury. Signal transduction by cAMP is highly compartmentalized in large part because of the formation of discrete, localized multimolecular signaling complexes by A-kinase anchoring proteins. Although the concept of cAMP compartmentation is well established, the function and identity of these compartments remain poorly understood in neurons. In this study, we provide evidence for a neuronal perinuclear cAMP compartment organized by the scaffold protein mAKAPα that is necessary and sufficient for the induction of neurite outgrowth in vitro and for the survival of retinal ganglion cells in vivo following optic nerve injury.


Subject(s)
Axon Guidance , Cyclic AMP/metabolism , Retinal Ganglion Cells/metabolism , Signal Transduction , A Kinase Anchor Proteins/metabolism , Animals , Axons/metabolism , Axons/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Fluorescence Resonance Energy Transfer , Male , Mice , Phosphoric Diester Hydrolases/metabolism , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology
13.
Dev Neurobiol ; 78(10): 998-1010, 2018 10.
Article in English | MEDLINE | ID: mdl-30027690

ABSTRACT

Retinal ganglion cells and other central nervous system neurons fail to regenerate after injury. Understanding the obstacles to survival and regeneration, and overcoming them, is key to preserving and restoring function. While comparisons in the cellular changes seen in these non-regenerative cells with those that do have intrinsic regenerative ability has yielded many candidate genes for regenerative therapies, complete visual recovery has not yet been achieved. Insights gained from neurodegenerative diseases, like glaucoma, underscore the importance of axonal transport of organelles, mRNA, and effector proteins in injury and disease. Targeting molecular motor networks, and their cargoes, may be necessary for realizing complete axonal regeneration and vision restoration.


Subject(s)
Axonal Transport/physiology , Glaucoma/metabolism , Nerve Regeneration/physiology , Neurodegenerative Diseases/metabolism , Neuroprotection/physiology , Retinal Ganglion Cells/physiology , Animals , Humans , Retinal Ganglion Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...