Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 117(31): 18810-18821, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32690710

ABSTRACT

In the central nervous system, glycogen-derived bioenergetic resources in astrocytes help promote tissue survival in response to focal neuronal stress. However, our understanding of the extent to which these resources are mobilized and utilized during neurodegeneration, especially in nearby regions that are not actively degenerating, remains incomplete. Here we modeled neurodegeneration in glaucoma, the world's leading cause of irreversible blindness, and measured how metabolites mobilize through astrocyte gap junctions composed of connexin 43 (Cx43). We elevated intraocular pressure in one eye and determined how astrocyte-derived metabolites in the contralateral optic projection responded. Remarkably, astrocyte networks expand and redistribute metabolites along distances even 10 mm in length, donating resources from the unstressed to the stressed projection in response to intraocular pressure elevation. While resource donation improves axon function and visual acuity in the directly stressed region, it renders the donating tissue susceptible to bioenergetic, structural, and physiological degradation. Intriguingly, when both projections are stressed in a WT animal, axon function and visual acuity equilibrate between the two projections even when each projection is stressed for a different length of time. This equilibration does not occur when Cx43 is not present. Thus, Cx43-mediated astrocyte metabolic networks serve as an endogenous mechanism used to mitigate bioenergetic stress and distribute the impact of neurodegenerative disease processes. Redistribution ultimately renders the donating optic nerve vulnerable to further metabolic stress, which could explain why local neurodegeneration does not remain confined, but eventually impacts healthy regions of the brain more broadly.


Subject(s)
Astrocytes , Glaucoma/metabolism , Neurodegenerative Diseases/metabolism , Animals , Astrocytes/metabolism , Astrocytes/physiology , Connexin 43/genetics , Connexin 43/metabolism , Female , Gap Junctions/metabolism , Intraocular Pressure/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
2.
Sci Rep ; 10(1): 8535, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444682

ABSTRACT

Glaucoma is a group of optic neuropathies associated with aging and sensitivity to intraocular pressure (IOP). Early progression involves retinal ganglion cell (RGC) axon dysfunction that precedes frank degeneration. Previously we demonstrated that p38 MAPK inhibition abates axonal dysfunction and slows degeneration in the inducible microbead occlusion model of glaucoma in rat. Here, we assessed the neuroprotective effect of topical eye delivery of the p38 MAPK inhibitor BIRB 796 in three models of glaucoma (microbead occlusion in rat and squirrel monkey and the genetic DBA/2 J mouse model) with distinct durations of IOP elevation. While BIRB 796 did not influence IOP, treatment over four weeks in rats prevented degradation of anterograde axonal transport to the superior colliculus and degeneration in the optic nerve. Treatment over months in the chronic DBA/2 J model and in the squirrel monkey model reduced expression and activation of p38 downstream targets in the retina and brain but did not rescue RGC axon transport or degeneration, suggesting the efficacy of BIRB 796 in preventing associated degeneration of the RGC projection depends on the duration of the experimental model. These results emphasize the importance of evaluating potential therapeutic compounds for neuroprotection in multiple models using elongated treatment paradigms for an accurate assessment of efficacy.


Subject(s)
Glaucoma/drug therapy , Naphthalenes/pharmacology , Neuroprotective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Axonal Transport/drug effects , Disease Models, Animal , Intraocular Pressure/drug effects , Male , Mice , Mice, Inbred DBA , Optic Nerve/drug effects , Optic Nerve/metabolism , Rats , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Saimiri
3.
Exp Eye Res ; 190: 107873, 2020 01.
Article in English | MEDLINE | ID: mdl-31734278

ABSTRACT

Glaucoma is an age-related neurodegenerative disease that is commonly associated with sensitivity to intraocular pressure. The disease selectively targets retinal ganglion cells (RGCs) and constituent axons. RGC axons are rich in voltage-gated sodium channels, which are essential for action potential initiation and regeneration. Here, we identified voltage-dependent sodium channel, NaV1.2, in the retina, examined how this channel contributes to RGC light responses, and monitored NaV1.2 mRNA and protein expression in the retina during progression of modeled glaucoma. We found NaV1.2 is predominately localized in ganglion cell intraretinal axons with dispersed expression in the outer and inner plexiform layers. We showed Phrixotoxin-3, a potent NaV1.2 channel blocker, significantly decreased RGC electrical activity in a dose-dependent manner with an IC50 of 40 nM. Finally, we found four weeks of raised intraocular pressure (30% above baseline) significantly increased NaV1.2 mRNA expression but reduced NaV1.2 protein level in the retina up to 57% (p < 0.001). Following prolonged intraocular pressure elevation, NaV1.2 protein expression particularly diminished at distal sections of ganglion cell intraretinal axons (p ≤ 0.01). Our results suggest NaV1.2 might be a therapeutic target during disease progression to maintain RGC excitability, preserving presynaptic connections through action potential backpropagation.


Subject(s)
Axons/metabolism , Intraocular Pressure/physiology , NAV1.2 Voltage-Gated Sodium Channel/metabolism , Ocular Hypertension/metabolism , Retinal Ganglion Cells/metabolism , Animals , Gene Expression Regulation/physiology , Male , Mice, Inbred C57BL , NAV1.2 Voltage-Gated Sodium Channel/genetics , RNA, Messenger/genetics , Tonometry, Ocular
4.
Sci Rep ; 9(1): 11572, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31399621

ABSTRACT

Glaucoma is a group of optic neuropathies associated with aging and sensitivity to intraocular pressure (IOP). The disease causes vision loss through the degeneration of retinal ganglion cell neurons and their axons in the optic nerve. Using an inducible model of glaucoma, we elevated IOP in the squirrel monkey (Saimiri boliviensis) using intracameral injection of 35 µm polystyrene microbeads and measured common pathogenic outcomes in the optic projection. A 42% elevation in IOP over 28 weeks reduced anterograde transport of fluorescently-labeled cholera toxin beta from retina to the lateral geniculate nucleus (60% decrease), and to the superior colliculus (49% decrease). Pressure also reduced survival of ganglion cellaxons in the optic nerve by 22%. The same elevation caused upregulation of proteins associated with glaucomatous neurodegeneration in the retina and optic nerve, including complement 1q, interleukin 6, and brain-derived neurotrophic factor. That axon degeneration in the nerve lagged deficits in anterograde transport is consistent with progression in rodent models, while the observed protein changes also occur in tissue from human glaucoma patients. Thus, microbead occlusion in a non-human primate with a visual system similar to our own represents an attractive model to investigate neurodegenerative mechanisms and therapeutic interventions for glaucoma.


Subject(s)
Disease Models, Animal , Glaucoma/physiopathology , Intraocular Pressure , Saimiri , Animals , Cell Survival , Complement C1q/analysis , Glaucoma/diagnosis , Glaucoma/pathology , Humans , Interleukin-6/analysis , Male , Optic Nerve/pathology , Optic Nerve/physiopathology , Saimiri/physiology
5.
Proc Natl Acad Sci U S A ; 115(10): E2393-E2402, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463759

ABSTRACT

Diseases of the brain involve early axon dysfunction that often precedes outright degeneration. Pruning of dendrites and their synapses represents a potential driver of axonopathy by reducing activity. Optic nerve degeneration in glaucoma, the world's leading cause of irreversible blindness, involves early stress to retinal ganglion cell (RGC) axons from sensitivity to intraocular pressure (IOP). This sensitivity also influences survival of RGC dendrites and excitatory synapses in the retina. Here we tested in individual RGCs identified by type the relationship between dendritic organization and axon signaling to light following modest, short-term elevations in pressure. We found dendritic pruning occurred early, by 2 wk of elevation, and independent of whether the RGC responded to light onset (ON cells) or offset (OFF cells). Pruning was similarly independent of ON and OFF in the DBA/2J mouse, a chronic glaucoma model. Paradoxically, all RGCs, even those with significant pruning, demonstrated a transient increase in axon firing in response to the preferred light stimulus that occurred on a backdrop of generally enhanced excitability. The increased response was not through conventional presynaptic signaling, but rather depended on voltage-sensitive sodium channels that increased transiently in the axon. Pruning, axon dysfunction, and deficits in visual acuity did not progress between 2 and 4 wk of elevation. These results suggest neurodegeneration in glaucoma involves an early axogenic response that counters IOP-related stress to excitatory dendritic architecture to slow progression and maintain signaling to the brain. Thus, short-term exposure to elevated IOP may precondition the neural system to further insult.


Subject(s)
Axons/physiology , Glaucoma/physiopathology , Retinal Ganglion Cells/physiology , Animals , Dendrites/physiology , Disease Progression , Glaucoma/pathology , Humans , Intraocular Pressure , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nerve Degeneration , Optic Nerve/physiopathology , Retinal Ganglion Cells/pathology
6.
Methods Mol Biol ; 1695: 23-39, 2018.
Article in English | MEDLINE | ID: mdl-29190015

ABSTRACT

Glaucoma is a common optic neuropathy that leads to vision loss through the degeneration of retinal ganglion cells (RGCs) and their axons. RGC degeneration in glaucoma is associated with sensitivity to intraocular pressure (IOP) and elevated IOP (also known as ocular hypertension) is the primary modifiable risk factor. Ocular hypertension is the primary characteristic of rodent models for glaucoma research. Intracameral injection of microbeads has evolved as a preferred method of IOP elevation in rodents, particularly in mice. Here, we outline the protocol and method for the Microbead Occlusion Model in mice. We highlight the importance of anesthesia choice and the utilization of glass micropipettes in combination with a micromanipulator and microsyringe pump for the successful execution of the model.


Subject(s)
Glaucoma/etiology , Microinjections/instrumentation , Ocular Hypertension/chemically induced , Animals , Disease Models, Animal , Glaucoma/physiopathology , Injections, Intraocular/instrumentation , Mice , Microspheres , Ocular Hypertension/complications , Ocular Hypertension/pathology , Retinal Ganglion Cells/pathology
7.
Methods Mol Biol ; 1618: 15-27, 2017.
Article in English | MEDLINE | ID: mdl-28523496

ABSTRACT

The visual system is comprised of many specialized cell types that are essential for relaying sensory information about an animal's surroundings to the brain. The cells present in ocular tissue are notoriously delicate, making it particularly challenging to section thin slices of unfixed tissue. Maintaining the morphology of the native tissue is crucial for accurate observations by either conventional staining techniques or in this instance matrix-assisted laser desorption ionization (MALDI IMS) or imaging using mass spectrometry. As vision loss is a significantly debilitating condition, studying molecular mechanisms involved in the process of vision loss is a critically important area of research.


Subject(s)
Optic Nerve/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Eye/physiopathology , Rodentia , Vision Disorders/physiopathology
8.
Front Neurosci ; 11: 45, 2017.
Article in English | MEDLINE | ID: mdl-28223915

ABSTRACT

Glaucoma is a group of optic neuropathies associated with aging and sensitivity to intraocular pressure (IOP). The disease is the leading cause of irreversible blindness worldwide. Early progression in glaucoma involves dysfunction of retinal ganglion cell (RGC) axons, which comprise the optic nerve. Deficits in anterograde transport along RGC axons to central visual structures precede outright degeneration, and preventing these deficits is efficacious at abating subsequent progression. HE3286 is a synthetic sterol derivative that has shown therapeutic promise in models of inflammatory disease and neurodegenerative disease. We examined the efficacy of HE3286 oral delivery in preventing loss of anterograde transport in an inducible model of glaucoma (microbead occlusion). Adult rats received HE3286 (20 or 100 mg/kg) or vehicle daily via oral gavage for 4 weeks. Microbead occlusion elevated IOP ~30% in all treatment groups, and elevation was not affected by HE3286 treatment. In the vehicle group, elevated IOP reduced anterograde axonal transport to the superior colliculus, the most distal site in the optic projection, by 43% (p = 0.003); HE3286 (100 mg/kg) prevented this reduction (p = 0.025). HE3286 increased brain-derived neurotrophic factor (BDNF) in the optic nerve head and retina, while decreasing inflammatory and pathogenic proteins associated with elevated IOP compared to vehicle treatment. Treatment with HE3286 also increased nuclear localization of the transcription factor NFκB in collicular and retinal neurons, but decreased NFκB in glial nuclei in the optic nerve head. Thus, HE3286 may have a neuroprotective influence in glaucoma, as well as other chronic neurodegenerations.

9.
Transl Vis Sci Technol ; 4(1): 1, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25599009

ABSTRACT

PURPOSE: We examined the efficacy of an extended-release drug delivery system, nanosponge (NS) encapsulated compounds, administered intravitreally to lower intraocular pressure (IOP) in mice. METHODS: Bilateral ocular hypertension was induced in mice by injecting microbeads into the anterior chamber. Hypertensive mice received NS loaded with ocular hypotensive drugs via intravitreal injection and IOP was monitored. Retinal deposition and retinal ganglion cell (RGC) uptake of Neuro-DiO were examined following intravitreal injection of Neuro-DiO-NS using confocal microscopy. RESULTS: Brimonidine-loaded NS lowered IOP 12% to 30% for up to 6 days (P < 0.02), whereas travoprost-NS lowered IOP 19% to 29% for up to 4 days (P < 0.02) compared to saline injection. Three bimatoprost NS were tested: a 400-nm NS and two 700-nm NS with amorphous (A-NS) or amorphous/crystalline (AC-NS) crosslinkers. A single injection of 400 nm NS lowered IOP 24% to 33% for up to 17 days compared to saline, while A-NS and AC-NS lowered IOP 22% to 32% and 18% to 26%, respectively, for up to 32 days (P < 0.046). Over time retinal deposition of Neuro-DiO increased from 19% to 71%; Neuro-DiO released from NS was internalized by RGCs. CONCLUSIONS: A single injection of NS can effectively deliver ocular hypotensive drugs in a linear and continuous manner for up to 32 days. Also, NS may be effective at targeting RGCs, the neurons that degenerate in glaucoma. TRANSLATIONAL RELEVANCE: Patient compliance is a major issue in glaucoma. The use of NS to deliver a controlled, sustained release of therapeutics could drastically reduce the number of patients that progress to vision loss in this disease.

10.
J Neurosci ; 34(46): 15369-81, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25392504

ABSTRACT

Progression of neurodegeneration in disease and injury is influenced by the response of individual neurons to stressful stimuli and whether this response includes mechanisms to counter declining function. Transient receptor potential (TRP) cation channels transduce a variety of disease-relevant stimuli and can mediate diverse stress-dependent changes in physiology, both presynaptic and postsynaptic. Recently, we demonstrated that knock-out or pharmacological inhibition of the TRP vanilloid-1 (TRPV1) capsaicin-sensitive subunit accelerates degeneration of retinal ganglion cell neurons and their axons with elevated ocular pressure, the critical stressor in the most common optic neuropathy, glaucoma. Here we probed the mechanism of the influence of TRPV1 on ganglion cell survival in mouse models of glaucoma. We found that induced elevations of ocular pressure increased TRPV1 in ganglion cells and its colocalization at excitatory synapses to their dendrites, whereas chronic elevation progressively increased ganglion cell Trpv1 mRNA. Enhanced TRPV1 expression in ganglion cells was transient and supported a reversal of the effect of TRPV1 on ganglion cells from hyperpolarizing to depolarizing, which was also transient. Short-term enhancement of TRPV1-mediated activity led to a delayed increase in axonal spontaneous excitation that was absent in ganglion cells from Trpv1(-/-) retina. In isolated ganglion cells, pharmacologically activated TRPV1 mobilized to discrete nodes along ganglion cell dendrites that corresponded to sites of elevated Ca(2+). These results suggest that TRPV1 may promote retinal ganglion cell survival through transient enhancement of local excitation and axonal activity in response to ocular stress.


Subject(s)
Retinal Ganglion Cells/physiology , Stress, Physiological/physiology , TRPV Cation Channels/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium/metabolism , Capsaicin/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Disease Models, Animal , Diterpenes/pharmacology , Dopamine/analogs & derivatives , Dopamine/pharmacology , Glaucoma/metabolism , Glaucoma/physiopathology , Intraocular Pressure/physiology , Mice , Mice, Knockout , Primary Cell Culture , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
11.
Glia ; 62(9): 1435-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24838827

ABSTRACT

Astrocytes provide metabolic, structural, and synaptic support to neurons in normal physiology and also contribute widely to pathogenic processes in response to stress or injury. Reactive astrocytes can undergo cytoskeletal reorganization and increase migration through changes in intracellular Ca(2+) mediated by a variety of potential modulators. Here we tested whether migration of isolated retinal astrocytes following mechanical injury (scratch wound) involves the transient receptor potential vanilloid-1 channel (TRPV1), which contributes to Ca(2+)-mediated cytoskeletal rearrangement and migration in other systems. Application of the TRPV1-specific antagonists, capsazepine (CPZ) or 5'-iodoresiniferatoxin (IRTX), slowed migration by as much as 44%, depending on concentration. In contrast, treatment with the TRPV1-specific agonists, capsaicin (CAP) or resiniferatoxin (RTX) produced only a slight acceleration over a range of concentrations. Chelation of extracellular Ca(2+) with EGTA (1 mM) slowed astrocyte migration by 35%. Ratiometric imaging indicated that scratch wound induced a sharp 20% rise in astrocyte Ca(2+) that dissipated with distance from the wound. Treatment with IRTX both slowed and dramatically reduced the scratch-induced Ca(2+) increase. Both CPZ and IRTX influenced astrocyte cytoskeletal organization, especially near the wound edge. Taken together, our results indicate that astrocyte mobilization in response to mechanical stress involves influx of extracellular Ca(2+) and cytoskeletal changes in part mediated by TRPV1 activation.


Subject(s)
Astrocytes/physiology , Cell Movement/physiology , Stress, Mechanical , TRPV Cation Channels/metabolism , Animals , Astrocytes/drug effects , Calcium/metabolism , Cell Culture Techniques , Cell Movement/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/physiology , Extracellular Space/metabolism , Intracellular Space/metabolism , Mice, Inbred C57BL , Rats, Sprague-Dawley , Retina/injuries , Retina/physiopathology , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors
12.
J Neurosci ; 34(9): 3161-70, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24573275

ABSTRACT

How neurons respond to stress in degenerative disease is of fundamental importance for identifying mechanisms of progression and new therapeutic targets. Members of the transient receptor potential (TRP) family of cation-selective ion channels are candidates for mediating stress signals, since different subunits transduce a variety of stimuli relevant in both normal and pathogenic physiology. We addressed this possibility for the TRP vanilloid-1 (TRPV1) subunit by comparing how the optic projection of Trpv1(-/-) mice and age-matched C57 controls responds to stress from elevated ocular pressure, the critical stressor in the most common optic neuropathy, glaucoma. Over a 5 week period of elevated pressure induced by microbead occlusion of ocular fluid, Trpv1(-/-) accelerated both degradation of axonal transport from retinal ganglion cells to the superior colliculus and degeneration of the axons themselves in the optic nerve. Ganglion cell body loss, which is normally later in progression, occurred in nasal sectors of Trpv1(-/-) but not C57 retina. Pharmacological antagonism of TRPV1 in rats similarly accelerated ganglion cell axonopathy. Elevated ocular pressure resulted in differences in spontaneous firing rate and action potential threshold current in Trpv1(-/-) ganglion cells compared with C57. In the absence of elevated pressure, ganglion cells in the two strains had similar firing patterns. Based on these data, we propose that TRPV1 may help neurons respond to disease-relevant stressors by enhancing activity necessary for axonal signaling.


Subject(s)
Nerve Degeneration , Optic Nerve Diseases , Retinal Ganglion Cells/pathology , TRPV Cation Channels/deficiency , Visual Pathways/pathology , Animals , Axons/pathology , Cholera Toxin , Disease Models, Animal , Functional Laterality , Intraocular Pressure/genetics , Male , Membrane Potentials/genetics , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/etiology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Ocular Hypertension/complications , Optic Nerve Diseases/etiology , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Patch-Clamp Techniques , Rats , Retinal Ganglion Cells/metabolism , Superior Colliculi/metabolism , Superior Colliculi/pathology , TRPV Cation Channels/genetics
13.
PLoS One ; 8(6): e65389, 2013.
Article in English | MEDLINE | ID: mdl-23755225

ABSTRACT

Oxidative stress has been implicated in neurodegenerative diseases, including glaucoma. However, due to the lack of clinically relevant models and expense of long-term testing, few studies have modeled antioxidant therapy for prevention of neurodegeneration. We investigated the contribution of oxidative stress to the pathogenesis of glaucoma in the DBA/2J mouse model of glaucoma. Similar to other neurodegenerative diseases, we observed lipid peroxidation and upregulation of oxidative stress-related mRNA and protein in DBA/2J retina. To test the role of oxidative stress in disease progression, we chose to deliver the naturally occurring, antioxidant α-lipoic acid (ALA) to DBA/2J mice in their diet. We used two paradigms for ALA delivery: an intervention paradigm in which DBA/2J mice at 6 months of age received ALA in order to intervene in glaucoma development, and a prevention paradigm in which DBA/2J mice were raised on a diet supplemented with ALA, with the goal of preventing glaucoma development. At 10 and 12 months of age (after 4 and 11 months of dietary ALA respectively), we measured changes in genes and proteins related to oxidative stress, retinal ganglion cell (RGC) number, axon transport, and axon number and integrity. Both ALA treatment paradigms showed increased antioxidant gene and protein expression, increased protection of RGCs and improved retrograde transport compared to control. Measures of lipid peroxidation, protein nitrosylation, and DNA oxidation in retina verified decreased oxidative stress in the prevention and intervention paradigms. These data demonstrate the utility of dietary therapy for reducing oxidative stress and improving RGC survival in glaucoma.


Subject(s)
Antioxidants/administration & dosage , Glaucoma/drug therapy , Retinal Ganglion Cells/drug effects , Thioctic Acid/administration & dosage , Administration, Oral , Animals , Axons/drug effects , Axons/physiology , Cell Death/drug effects , DNA Damage , Dietary Supplements , Drug Evaluation, Preclinical , Gene Expression , Glaucoma/pathology , Intraocular Pressure/drug effects , Lipid Peroxidation , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nerve Degeneration/prevention & control , Nitric Oxide Synthase Type II/metabolism , Oxidation-Reduction , Oxidative Stress , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/physiology , Treatment Outcome , Up-Regulation
14.
Mol Vis ; 19: 581-92, 2013.
Article in English | MEDLINE | ID: mdl-23559852

ABSTRACT

PURPOSE: To develop a method for generating high spatial resolution (10 µm) matrix-assisted laser desorption ionization (MALDI) images of lipids in rodent optic nerve tissue. METHODS: Ice-embedded optic nerve tissue from rats and mice were cryosectioned across the coronal and sagittal axes of the nerve fiber. Sections were thaw mounted on gold-coated MALDI plates and were washed with ammonium acetate to remove biologic salts before being coated in 2,5-dihydroxybenzoic acid by sublimation. MALDI images were generated in positive and negative ion modes at 10 µm spatial resolution. Lipid identification was performed with a high mass resolution Fourier transform ion cyclotron resonance mass spectrometer. RESULTS: Several lipid species were observed with high signal intensity in MALDI images of optic nerve tissue. Several lipids were localized to specific structures including in the meninges surrounding the optic nerve and in the central neuronal tissue. Specifically, phosphatidylcholine species were observed throughout the nerve tissue in positive ion mode while sulfatide species were observed in high abundance in the meninges surrounding the optic nerve in negative ion mode. Accurate mass measurements and fragmentation using sustained off-resonance irradiation with a high mass resolution Fourier transform ion cyclotron resonance mass spectrometer instrument allowed for identification of lipid species present in the small structure of the optic nerve directly from tissue sections. CONCLUSIONS: An optimized sample preparation method provides excellent sensitivity for lipid species present within optic nerve tissue. This allowed the laser spot size and fluence to be reduced to obtain a high spatial resolution of 10 µm. This new imaging modality can now be applied to determine spatial and molecular changes in optic nerve tissue with disease.


Subject(s)
Lipids/analysis , Lipids/chemistry , Optic Nerve/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Mice , Mice, Inbred C57BL , Optical Imaging , Rats , Rats, Inbred BN
15.
Mol Neurodegener ; 6(1): 4, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21232114

ABSTRACT

BACKGROUND: Brimonidine is a common drug for lowering ocular pressure and may directly protect retinal ganglion cells in glaucoma. The disease involves early loss of retinal ganglion cell transport to brain targets followed by axonal and somatic degeneration. We examined whether brimonidine preserves ganglion cell axonal transport and abates degeneration in rats with elevated ocular pressure induced by laser cauterization of the episcleral veins. RESULTS: Ocular pressure was elevated unilaterally by 90% for a period of 8 weeks post- cauterization. During this time, brimonidine (1mg/kg/day) or vehicle (phosphate-buffered saline) was delivered systemically and continuously via subcutaneous pump. Animals received bilateral intravitreal injections of fluorescent cholera toxin subunit ß (CTB) two days before sacrifice to assess anterograde transport. In retinas from the vehicle group, elevated pressure induced a 44% decrease in the fraction of ganglion cells with intact uptake of CTB and a 14-42% reduction in the number of immuno-labelled ganglion cell bodies, with the worst loss occurring nasally. Elevated pressure also caused a 33% loss of ganglion cell axons in vehicle optic nerves and a 70% decrease in CTB transport to the superior colliculus. Each of these components of ganglion cell degeneration was either prevented or significantly reduced in the brimonidine treatment group. CONCLUSIONS: Continuous and systemic treatment with brimonidine by subcutaneous injection significantly improved retinal ganglion cell survival with exposure to elevated ocular pressure. This effect was most striking in the nasal region of the retina. Brimonidine treatment also preserved ganglion cell axon morphology, sampling density and total number in the optic nerve with elevated pressure. Consistent with improved outcome in the optic projection, brimonidine also significantly reduced the deficits in axonal transport to the superior colliculus associated with elevated ocular pressure. As transport deficits to and from retinal ganglion cell projection targets in the brain are relevant to the progression of glaucoma, the ability of brimonidine to preserve optic nerve axons and active transport suggests its neuroprotective effects are relevant not only at the cell body, but throughout the entire optic projection.

16.
Invest Ophthalmol Vis Sci ; 52(1): 504-18, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20847120

ABSTRACT

PURPOSE: In glaucoma, the optic nerve head (ONH) is the principal site of initial axonal injury, and elevated intraocular pressure (IOP) is the predominant risk factor. However, the initial responses of the ONH to elevated IOP are unknown. Here the authors use a rat glaucoma model to characterize ONH gene expression changes associated with early optic nerve injury. METHODS: Unilateral IOP elevation was produced in rats by episcleral vein injection of hypertonic saline. ONH mRNA was extracted, and retrobulbar optic nerve cross-sections were graded for axonal degeneration. Gene expression was determined by microarray and quantitative PCR (QPCR) analysis. Significantly altered gene expression was determined by multiclass analysis and ANOVA. DAVID gene ontology determined the functional categories of significantly affected genes. RESULTS: The Early Injury group consisted of ONH from eyes with <15% axon degeneration. By array analysis, 877 genes were significantly regulated in this group. The most significant upregulated gene categories were cell cycle, cytoskeleton, and immune system process, whereas the downregulated categories included glucose and lipid metabolism. QPCR confirmed the upregulation of cell cycle-associated genes and leukemia inhibitory factor (Lif) and revealed alterations in expression of other IL-6-type cytokines and Jak-Stat signaling pathway components, including increased expression of IL-6 (1553%). In contrast, astrocytic glial fibrillary acidic protein (Gfap) message levels were unaltered, and other astrocytic markers were significantly downregulated. Microglial activation and vascular-associated gene responses were identified. CONCLUSIONS: Cell proliferation and IL-6-type cytokine gene expression, rather than astrocyte hypertrophy, characterize early pressure-induced ONH injury.


Subject(s)
Cell Proliferation , Gene Expression Regulation/physiology , Glaucoma/genetics , Interleukin-6/genetics , Optic Disk/metabolism , Optic Nerve Injuries/genetics , Signal Transduction/physiology , Animals , Axons/metabolism , Axons/pathology , Disease Models, Animal , Glaucoma/pathology , Intraocular Pressure , Male , Microarray Analysis , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Optic Disk/pathology , Optic Nerve Injuries/pathology , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Rats, Inbred BN
17.
J Neurosci ; 28(11): 2735-44, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18337403

ABSTRACT

Glaucoma is characterized by retinal ganglion cell (RGC) pathology and a progressive loss of vision. Previous studies suggest RGC death is responsible for vision loss in glaucoma, yet evidence from other neurodegenerative diseases suggests axonal degeneration, in the absence of neuronal loss, can significantly affect neuronal function. To characterize RGC degeneration in the DBA/2 mouse model of glaucoma, we quantified RGCs in mice of various ages using neuronal-specific nuclear protein (NeuN) immunolabeling, retrograde labeling, and optic nerve axon counts. Surprisingly, the number of NeuN-labeled RGCs did not decline significantly until 18 months of age, at which time a significant decrease in RGC somal size was also observed. Axon dysfunction and degeneration occurred before loss of NeuN-positive RGCs, because significant declines in RGC number assayed by retrograde tracers and axon counts were observed at 13 months. To examine whether axonal dysfunction/degeneration affected gene expression in RGC axons or somas, NeuN and neurofilament-heavy (NF-H) immunolabeling was performed along with quantitative reverse transcription-PCR for RGC-specific genes in retinas of aged DBA/2 mice. Although these mice had similar numbers of NeuN-positive RGCs, the expression of neurofilament light, Brn-3b, and Sncg mRNA varied; this variation in RGC-specific gene expression was correlated with the appearance of NF-H immunoreactive RGC axons. Together, these data support a progression of RGC degeneration in this model of glaucoma, beginning with loss of retrograde label, where axon dysfunction and degeneration precede neuronal loss. This progression of degeneration suggests a need to examine the RGC axon as a locus of pathology in glaucoma.


Subject(s)
Disease Models, Animal , Glaucoma/pathology , Nerve Degeneration/pathology , Neurons/pathology , Retinal Ganglion Cells/pathology , Animals , Cell Count/methods , Cell Death/physiology , Disease Progression , Glaucoma/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nerve Degeneration/genetics , Neurons/physiology , Retinal Ganglion Cells/physiology
18.
BMC Neurosci ; 5: 51, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15579199

ABSTRACT

BACKGROUND: Ischemia within the optic nerve head (ONH) may contribute to retinal ganglion cell (RGC) loss in primary open angle glaucoma (POAG). Ischemia has been reported to increase neurotrophin and high affinity Trk receptor expression by CNS neurons and glial cells. We have previously demonstrated neurotrophin and Trk expression within the lamina cribrosa (LC) region of the ONH. To determine if ischemia alters neurotrophin and Trk protein expression in cells from the human LC, cultured LC cells and ONH astrocytes were exposed to 48 hours of oxygen-glucose deprivation (OGD). Also cells were exposed to 48 hours of OGD followed by 24 hours of recovery in normal growth conditions. Cell number, neurotrophin and Trk receptor protein expression, neurotrophin secretion, and Trk receptor activation were examined. RESULTS: Cell number was estimated using an assay for cell metabolism following 24, 48 and 72 hours of OGD. A statistically significant decrease in LC and ONH astrocyte cell number did not occur until 72 hours of OGD, therefore cellular protein and conditioned media were collected at 48 hours OGD. Protein expression of NGF, BDNF and NT-3 by LC cells and ONH astrocytes increased following OGD, as did NGF secretion. Recovery from OGD increased BDNF protein expression in LC cells. In ONH astrocytes, recovery from OGD increased NGF protein expression, and decreased BDNF secretion. Trk A expression and activation in LC cells was increased following OGD while expression and activation of all other Trk receptors was decreased. A similar increase in Trk A expression and activation was observed in ONH astrocytes following recovery from OGD. CONCLUSIONS: In vitro conditions that mimic ischemia increase the expression and secretion of neurotrophins by cells from the ONH. Increased Trk A expression and activation in LC cells following OGD and in ONH astrocytes following recovery from OGD suggest autocrine/paracrine neurotrophin signaling could be a response to ONH ischemia in POAG. Also, the increase in NGF, BDNF and NT-3 protein expression and NGF secretion following OGD also suggest LC cells and ONH astrocytes may be a paracrine source of neurotrophins for RGCs.


Subject(s)
Nerve Growth Factors/metabolism , Optic Disk/cytology , Receptors, Nerve Growth Factor/metabolism , Cell Count , Cell Hypoxia , Cells, Cultured , Glucose/physiology , Humans , Optic Disk/metabolism , Optic Neuropathy, Ischemic/metabolism , Phosphorylation
19.
Mol Vis ; 10: 289-96, 2004 Apr 19.
Article in English | MEDLINE | ID: mdl-15105791

ABSTRACT

PURPOSE: Glaucoma is the number one cause of preventable blindness in the United States. The lamina cribrosa (LC) region of the optic nerve head (ONH) is a major site of injury in glaucomatous optic neuropathy. Neurotrophins (NTs), which include NGF, BDNF, NT-3, and NT-4, are growth factors involved in the development and support of neurons and in non-neuronal interactions. Cells within the human LC express high affinity tyrosine kinase receptors (Trks) for NTs. The purpose of this study was to determine if exogenous NTs cause (a) phosphorylation of Trk receptors in LC cells and ONH astrocytes and (b) cell proliferation and/or secretion of NTs by LC cells and ONH astrocytes. METHODS: Trk phosphorylation in response to exogenous NGF, BDNF, NT-3, and NT-4 treatment was studied in LC cells and ONH astrocytes using immunoprecipitation and Western blotting. Cell number was assayed following treatment with exogenous NTs or the Trk phosphorylation inhibitor compound K-252a. Secretion of NTs following exogenous administration of NTs was determined using immunoassays. RESULTS: LC cells and ONH astrocytes express Trk receptors that are phosphorylated in response to exogenous NTs. Autocrine/paracrine signaling was also evident by Trk phosphorylation in the absence of exogenous NT treatment. ONH astrocyte cell number increased following exogenous treatment with each NT. LC cell number increased following exogenous NGF or NT-3 treatment only. Treatment with the Trk phosphorylation inhibitor K-252a decreased both LC and ONH astrocyte cell number. Exogenous NT treatment increased the secretion of NGF by LC cells and ONH astrocytes. BDNF secretion by LC cells and ONH astrocytes was decreased by exogenous NT treatment. CONCLUSIONS: LC cells and ONH astrocytes express functional Trk receptors that can be activated in response to exogenous NTs. The activation of Trk receptors expressed by LC cells and ONH astrocytes in the absence of exogenous NT treatment suggests autocrine/paracrine NT signaling may occur within the ONH. Neurotrophin signaling in LC cells and ONH astrocytes may regulate cell number and/or NT secretion within the LC region of the ONH.


Subject(s)
Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Optic Disk/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Actins/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Carbazoles/pharmacology , Cell Count , Cell Division/drug effects , Collagen/metabolism , Elastin/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein/metabolism , Humans , Indole Alkaloids , Nerve Growth Factor/metabolism , Neural Cell Adhesion Molecules/metabolism , Optic Disk/cytology , Optic Disk/metabolism , Phosphorylation
20.
Mol Vis ; 9: 249-56, 2003 Jun 16.
Article in English | MEDLINE | ID: mdl-12813408

ABSTRACT

PURPOSE: Glial cell-line derived neurotrophic factor (GDNF) is a distant member of the TGFbeta family of growth factors and has wide ranging effects within the central nervous system. In the present study we profile the expression of GDNF and its receptor complex (Ret and GFRalpha-1) in cells isolated from the human optic nerve head (ONH). METHODS: Lamina cribrosa (LC) cells and ONH astrocytes were used from normal donors of various ages. Total RNA was isolated and subjected to reverse transcriptase-polymerase chain reaction (RT-PCR) to examine mRNA expression of GDNF, Ret, and GFRalpha-1. Western immunoblotting and immunohistochemistry was used to study protein expression of GDNF and GDNF receptor complex proteins in cultured ONH cells. An immunoassay system (ELISA) was used to examine secretion of GDNF by ONH cells. Cell proliferation was examined following exogenous administration of GDNF. RESULTS: Lamina cribrosa cells, ONH astrocytes, and LC tissues expressed messenger RNA for GDNF, Ret and GFRalpha-1. Lamina cribrosa cells and ONH astrocytes also expressed protein for GDNF, Ret, and GFRalpha-1. Secretion of GDNF by either cell type was not detected. Exogenous GDNF caused a significant increase in cell proliferation of LC cells but not ONH astrocytes. CONCLUSIONS: Cells from the human lamina cribrosa express mRNA and protein for GDNF and its receptor complex. LC cells proliferate in response to exogenous GDNF. The potential for autocrine and/or paracrine GDNF signaling thus exists within the lamina cribrosa, a tissue involved in glaucoma pathogenesis.


Subject(s)
Astrocytes/metabolism , Nerve Growth Factors/genetics , Oncogene Proteins/genetics , Optic Disk/cytology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Astrocytes/cytology , Blotting, Western , Cell Division/drug effects , Cells, Cultured , Child , Child, Preschool , Fluorescent Antibody Technique, Indirect , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Infant , Infant, Newborn , Middle Aged , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , RNA/isolation & purification , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...