Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 6: 29523, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27389831

ABSTRACT

Abnormal α-synuclein (α-syn) accumulation in the CNS may underlie neuronal cell and synaptic dysfunction leading to motor and cognitive deficits in synucleinopathies including Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB). Multiple groups demonstrated α-syn accumulation in CNS accessory structures, including the eyes and olfactory terminals, as well as in peripheral organs of Parkinsonian patients. Retinal imaging studies of mice overexpressing fused α-syn::GFP were conducted to evaluate the presence and progression of retinal pathology in a PD/DLB transgenic mouse model. Bright-field image retinal maps and fluorescent images were acquired at 1-month intervals for 3 months. Retinal imaging revealed the accumulation of GFP-tagged α-syn in retinal ganglion cell layer and in the edges of arterial blood vessels in the transgenic mice. Double labeling studies confirmed that the α-syn::GFP-positive cells were retinal ganglion cells containing α-syn. Accumulation of α-syn persisted in the same cells and increased with age. Accumulation of α-syn::GFP was reduced by immunization with single chain antibodies against α-syn. In conclusion, longitudinal live imaging of the retina in the PDGF-α-syn::GFP mice might represent a useful, non-invasive tool to monitor the fate of α-syn accumulation in the CNS and to evaluate the therapeutic effects of compounds targeting α-syn.


Subject(s)
Green Fluorescent Proteins/metabolism , Lewy Body Disease/diagnostic imaging , Parkinson Disease/diagnostic imaging , Retina/diagnostic imaging , Single-Chain Antibodies/administration & dosage , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Disease Progression , Green Fluorescent Proteins/genetics , Humans , Immunotherapy , Lewy Body Disease/metabolism , Lewy Body Disease/therapy , Male , Mice , Mice, Transgenic , Optical Imaging/instrumentation , Parkinson Disease/metabolism , Parkinson Disease/therapy , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism , Single-Chain Antibodies/pharmacology , alpha-Synuclein/drug effects , alpha-Synuclein/genetics
2.
Neurobiol Aging ; 36(5): 1969-81, 2015 May.
Article in English | MEDLINE | ID: mdl-25772060

ABSTRACT

Long-term dietary supplementation with resveratrol protects against cardiovascular disease, osteoporesis, and metabolic decline. This study determined how long-term dietary resveratrol treatment protects against retinal ganglion cell (RGC) dendrite loss after optic nerve injury and alters the resolution of the unfolded protein response. Associated changes in markers of endoplasmic reticulum stress in RGCs also were investigated. Young-adult Thy1-yellow fluorescent protein (YFP) and C57BL/6 mice received either control diet or diet containing resveratrol for approximately 1 year. Both groups then received optic nerve crush (ONC). Fluorescent RGC dendrites in the Thy1-YFP mice were imaged weekly for 4 weeks after ONC. There was progressive loss of dendrite length in all RGC types within the mice that received control diet. Resveratrol delayed loss of dendrite complexity and complete dendrite loss for most RGC types. However, there were variations in the rate of retraction among different RGC types. Three weeks after ONC, cytoplasmic binding immunoglobulin protein (BiP) suppression observed in control diet ganglion cell layer neurons was reversed in mice that received resveratrol, nuclear C/EBP homologous protein (CHOP) was near baseline in control diet eyes but was moderately increased by resveratrol; and increased nuclear X-box-binding protein-1 (XBP-1) observed in control diet eyes was reduced in eyes that received resveratrol to the same level as in control diet uncrushed eyes. These results indicate that protection of dendrites by resveratrol after ONC differs among RGC types and suggest that alterations in long-term expression of binding immunoglobulin protein, CHOP, and XBP-1 may contribute to the resveratrol-mediated protection of RGC dendrites after ONC.


Subject(s)
Dendrites/metabolism , Dendrites/pathology , Dietary Supplements , Optic Nerve Injuries/pathology , Protein Unfolding/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Stilbenes/administration & dosage , Stilbenes/pharmacology , Animals , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Heat-Shock Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Regulatory Factor X Transcription Factors , Resveratrol , Retinal Ganglion Cells/cytology , Time Factors , Transcription Factor CHOP/metabolism , Transcription Factors/metabolism , X-Box Binding Protein 1
3.
Invest Ophthalmol Vis Sci ; 56(3): 1789-804, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25634981

ABSTRACT

PURPOSE: To determine whether brimonidine protects against the retraction and loss of retinal ganglion cell (RGC) dendrites after optic nerve crush (ONC). METHODS: Fluorescent RGCs of mice expressing yellow fluorescent protein (YFP) under the control of the Thy-1 promoter (Thy1-YFP mice) were imaged in vivo and assigned to one of six groups according to dendrite structure. The mice then received brimonidine every other day starting 2 days before, or 2 or 6 days after, unilateral ONC. Control animals received vehicle every other day starting 2 days before ONC. Control animals received vehicle every other day starting 2 days before ONC. Total dendrite length, dendrite branching complexity, and the time until complete loss of dendrites were assessed weekly for 4 weeks. RESULTS: Overall, brimonidine treatment significantly slowed the complete loss of RGC dendrites and significantly slowed the reduction of total dendrite length and branching complexity. Separate analysis of each RGC group showed brimonidine significantly delayed the time until complete loss of dendrites in four of the RGC groups. These delays generally were similar when treatment started either 2 days before or 2 days after ONC, but were smaller or absent when treatment started 6 days after ONC Protection against loss of total dendrite length and loss of branching complexity was observed in three of the RGC groups. In two of these RGC groups, protective effects persisted until the end of the study. CONCLUSIONS: Brimonidine protects many RGC types against dendrite retraction, loss of branching complexity, and complete loss of dendrites following ONC. However, the pattern and magnitude of this protection differs substantially among different RGC types. These results indicate that requirements for RGC-protective therapies following optic nerve injury may differ among RGC types.


Subject(s)
Cytoprotection/drug effects , Dendrites/drug effects , Nerve Crush , Optic Nerve Injuries/pathology , Quinoxalines/pharmacology , Retinal Ganglion Cells/drug effects , Animals , Brimonidine Tartrate , Dendrites/pathology , Female , Male , Mice , Mice, Inbred Strains , Microscopy, Fluorescence , Retinal Ganglion Cells/pathology
4.
PLoS One ; 8(8): e65966, 2013.
Article in English | MEDLINE | ID: mdl-23940507

ABSTRACT

Thy-1 is a cell surface protein that is expressed during the differentiation of retinal ganglion cells (RGCs). Optic nerve injury induces progressive loss in the number of RGCs expressing Thy-1. The rate of this loss is fastest during the first week after optic nerve injury and slower in subsequent weeks. This study was undertaken to determine whether oral treatment with a water-soluble N-hydroxy-2,2,6,6-tetramethylpiperidine derivative (OT-440) protects against loss of Thy-1 promoter activation following optic nerve crush and whether this effect targets the earlier quick phase or the later slow phase. The retina of mice expressing cyan fluorescent protein under control of the Thy-1 promoter (Thy1-CFP mice) was imaged using a blue-light confocal scanning laser ophthalmoscope (bCSLO). These mice then received oral OT-440 prepared in cream cheese or dissolved in water, or plain vehicle, for two weeks and were imaged again prior to unilateral optic nerve crush. Treatments and weekly imaging continued for four more weeks. Fluorescent neurons were counted in the same defined retinal areas imaged at each time point in a masked fashion. When the counts at each time point were directly compared, the numbers of fluorescent cells at each time point were greater in the animals that received OT-440 in cream cheese by 8%, 27%, 52% and 60% than in corresponding control animals at 1, 2, 3 and 4 weeks after optic nerve crush. Similar results were obtained when the vehicle was water. Rate analysis indicated the protective effect of OT-440 was greatest during the first two weeks and was maintained in the second two weeks after crush for both the cream cheese vehicle study and water vehicle study. Because most of the fluorescent cells detected by bCSLO are RGCs, these findings suggest that oral OT-440 can either protect against or delay early degenerative responses occurring in RGCs following optic nerve injury.


Subject(s)
Cell Differentiation/drug effects , Hydroxylamine/chemistry , Hydroxylamine/pharmacology , Optic Nerve Injuries/drug therapy , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Administration, Oral , Animals , Body Weight/drug effects , Female , Hydroxylamine/administration & dosage , Hydroxylamine/therapeutic use , Male , Mice , Microscopy, Confocal , Nerve Crush , Retinal Ganglion Cells/metabolism , Thy-1 Antigens/metabolism
5.
BMC Ophthalmol ; 13(1): 26, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23805828

ABSTRACT

BACKGROUND: The loss of RGCs expressing Thy-1 after optic nerve injury has an initial phase of rapid decline followed by a longer phase with slower reduction rate. This study used longitudinal retinal imaging of mice expressing cyan fluorescent protein under control of the Thy-1 promoter (Thy1-CFP mice) to determine how the α2-adrenergic agonist brimonidine influences loss of Thy1 promoter activation. METHODS: Baseline images of the fluorescent retinal neurons in 30 Thy1-CFP mice were obtained using a modified confocal scanning laser ophthalmoscope. Next, brimonidine (100 ug/kg, IP) was administered either one time immediately after optic nerve crush, or immediately after optic nerve crush and then every 2 days for four weeks. A control group received a single saline injection immediately after optic nerve crush. All animals were imaged weekly for four weeks after optic nerve crush. Loss of fluorescent retinal neurons within specific retinal areas was determined by counting. RESULTS: At one week after optic nerve crush, the proportion of fluorescent retinal neurons retaining fluorescence was 44±7% of baseline in control mice, 51±6% after one brimonidine treatment, and 55±6% after brimonidine treatment every other day (P<0.05 for both brimonidine treatment groups compared to the control group). Subsequently, the number of fluorescent retinal neurons in the group that received one treatment differed insignificantly from the control group. In contrast, the number of fluorescent retinal neurons in the group that received repeated brimonidine treatments was greater than the control group by 28% at two weeks after crush and by 32% at three weeks after crush (P<0.05 at both time points). Rate analysis showed that brimonidine slowed the initial rate of fluorescent cell decline in the animals that received multiple treatments (P<0.05). Differences in the rate of loss among the treatment groups were insignificant after the second week. CONCLUSION: Repeated brimonidine treatments protect against loss of fluorescence within fluorescent retinal neurons of Thy1-CFP mice after optic nerve crush. As most of fluorescent retinal neurons in this system are RGCs, these findings indicate that repeated brimonidine treatments may protect RGC health following optic nerve crush.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/therapeutic use , Nerve Crush , Optic Nerve Injuries/drug therapy , Promoter Regions, Genetic , Protective Agents/therapeutic use , Quinoxalines/therapeutic use , Thy-1 Antigens/physiology , Analysis of Variance , Animals , Brimonidine Tartrate , Cell Count , Disease Models, Animal , Female , Longitudinal Studies , Male , Mice , Microscopy, Fluorescence , Optic Nerve Injuries/genetics , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/drug effects
6.
Mol Vis ; 16: 1331-42, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20664796

ABSTRACT

PURPOSE: The goal of this study is to determine whether increased optic atrophy type 1 (OPA1) expression protects against retinal ganglion cell (RGC) death in glaucomatous DBA/2J mice. METHODS: Intraocular pressure in DBA/2J mice was measured, and pre-glaucomatous DBA/2J mice eyes were transfected with recombinant adeno-associated virus serotype 2 (AAV2) constructs including AAV2-wild type (WT) mOPA1 for two months. Increased OPA1 expression was confirmed by western blotting and RGC survival was assessed by retrograde labeling with FluoroGold. In addition, apoptotic cell death and mitochondrial structure were determined in AAV2-WT mOPA1-transfected differentiated RGC-5 cells exposed to elevated hydrostatic pressure (30 mmHg) for three days. RESULTS: WT AAV2-mOPA1 transfection significantly increased 90 kDa and 80 kDa OPA1 isoforms in the retina of glaucomatous DBA/2J mice. OPA1 immunoreactivity was increased in the inner nuclear layer, inner plexiform layer, and ganglion cell layer in nine month-old glaucomatous DBA/2J mice transfected with AAV2-WT mOPA1. Overexpression of OPA1 significantly increased RGC survival at two months after AAV2-WT mOPA1 transfection, and decreased activation of both astroglia and microglia in the retina of glaucomatous DBA/2J mice. Also, overexpression of OPA1 in differentiated RGC-5 cells resulted in less apoptotic cell death and blocked mitochondrial fission following elevated hydrostatic pressure. CONCLUSIONS: OPA1 can directly modulate RGC survival, and increasing OPA1 expression may protect against RGC death in glaucomatous optic neuropathy.


Subject(s)
Cytoprotection , GTP Phosphohydrolases/metabolism , Glaucoma/enzymology , Glaucoma/pathology , Retinal Ganglion Cells/enzymology , Animals , Apoptosis , Astrocytes/enzymology , Astrocytes/pathology , Cell Survival , Dependovirus/metabolism , Disease Models, Animal , Glaucoma/physiopathology , Green Fluorescent Proteins/metabolism , Intraocular Pressure/physiology , Mice , Mice, Inbred C57BL , Microglia/enzymology , Microglia/pathology , Mitochondria/metabolism , Retinal Ganglion Cells/pathology , Transfection
7.
Proc Natl Acad Sci U S A ; 106(21): 8760-5, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19433784

ABSTRACT

Many ocular pathologies, including retinopathy of prematurity (ROP), diabetic retinopathy, and age-related macular degeneration, result in vision loss because of aberrant neoangiogenesis. A common feature of these conditions is the presence of hypoxic areas and overexpression of the proangiogenic vascular endothelial growth factor (VEGF). The prevailing current treatment, laser ablation of the retina, is destructive and only partially effective. Preventive and less destructive therapies are much more desirable. Here, we show that mice lacking c-Jun N-terminal kinase 1 (JNK1) exhibit reduced pathological angiogenesis and lower levels of retinal VEGF production in a murine model of ROP. We found that hypoxia induces JNK activation and regulates VEGF expression by enhancing the binding of phospho-c-Jun to the VEGF promoter. Intravitreal injection of a specific JNK inhibitor decreases retinal VEGF expression and reduces pathological retinal neovascularization without obvious side effects. These results strongly suggest that JNK1 plays a key role in retinal neoangiogenesis and that it represents a new pharmacological target for treatment of diseases where excessive neoangiogenesis is the underlying pathology.


Subject(s)
Hypoxia/metabolism , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/metabolism , Retinal Diseases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation/drug effects , Hypoxia/genetics , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mitogen-Activated Protein Kinase 8/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Protein Kinase Inhibitors/pharmacology , Retinal Diseases/genetics , Retinal Diseases/pathology , Transcription, Genetic/genetics , Vascular Endothelial Growth Factor A/genetics
8.
Invest Ophthalmol Vis Sci ; 50(2): 707-16, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18936150

ABSTRACT

PURPOSE: To determine whether intraocular pressure (IOP) elevation alters OPA1 expression and triggers OPA1 release, as well as whether the uncompetitive N-methyl-d-aspartate (NMDA) glutamate receptor antagonist memantine blocks OPA1 release and subsequent apoptotic cell death in glaucomatous DBA/2J mouse retina. METHODS: Preglaucomatous DBA/2J mice received memantine (5 mg/kg, intraperitoneal injection, twice daily for 3 months) and IOP in the eyes was measured monthly. RGC loss was counted after FluoroGold labeling. OPA1, Dnm1, Bcl-2, and Bax mRNA were measured by qPCR. OPA1 protein was assessed by immunohistochemistry and Western blot. Apoptotic cell death was assessed by TUNEL staining. RESULTS: Memantine treatment significantly increased RGC survival in glaucomatous DBA/2J mice and increased the 75-kDa OPA1 isoform, but did not alter the 80- and 90-kDa isoforms. The isoforms of OPA1 were significantly increased in the cytosol of the vehicle-treated glaucomatous retinas but were significantly decreased in memantine-treated glaucomatous retinas. OPA1 immunoreactivity was decreased in the photoreceptors of both vehicle- and memantine-treated glaucomatous retinas, but was increased in the outer plexiform layer of only the memantine-treated glaucomatous retinas. Memantine blocked apoptotic cell death in the GCL, increased Bcl-2 gene expression, and decreased Bax gene expression. CONCLUSIONS: OPA1 release from mitochondria in glaucomatous mouse retina is inhibited by blockade of glutamate receptor activation. Because this OPA1 effect was accompanied by increased Bcl-2 expression, decreased Bax expression, and apoptosis blockade, glutamate receptor activation in the glaucomatous retina may involve a distinct mitochondria-mediated cell death pathway.


Subject(s)
Apoptosis/drug effects , Cytochromes c/metabolism , GTP Phosphohydrolases/metabolism , Glaucoma/prevention & control , Memantine/pharmacology , Mitochondria/drug effects , Retinal Diseases/prevention & control , Animals , Blotting, Western , Cell Survival , Cytochromes c/genetics , Dynamin I/genetics , Excitatory Amino Acid Antagonists/pharmacology , Female , GTP Phosphohydrolases/genetics , Gene Expression , Glaucoma/genetics , Glaucoma/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Injections, Intraperitoneal , Intraocular Pressure , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mitochondria/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Ganglion Cells/drug effects , bcl-2-Associated X Protein/genetics
9.
Invest Ophthalmol Vis Sci ; 49(11): 4903-11, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18469184

ABSTRACT

PURPOSE: To determine whether elevation of intraocular pressure (IOP) triggers mitochondrial fission and ultrastructural changes and alters optic atrophy type 1 (OPA1) expression and distribution in the optic nerve (ON) of glaucomatous DBA/2J mice. METHODS: IOP in the eyes of DBA/2J mice was measured, and mitochondrial structural changes were assessed by conventional electron microscopy (EM) and EM tomography. Cytochrome c oxidase IV subunit 1 (COX), OPA1, and Dnm1, a rat homologue of dynamin-related protein-1, mRNA were measured by quantitative (q)PCR. COX and OPA1 protein distribution was assessed by immunocytochemistry and Western blot. RESULTS: Excavation of the optic nerve head (ONH), axon loss, and COX reduction were evident in 10-month-old glaucomatous ONHs of eyes with >20 mm Hg IOP elevation. EM analysis showed mitochondrial fission, matrix swelling, substantially reduced cristae volume, and abnormal cristae depletion in 10-month-old glaucomatous ONH axons. The mean length of mitochondrial cross section in these axons decreased from 858.2 +/- 515.3 nm in 3-month-old mice to 583.3 +/- 298.6 nm in 10-month-old glaucomatous mice (P < 0.001). Moderate reductions of COX mRNA were observed in the 10-month-old DBA/2J mice's ONHs. Larger reductions of OPA1 immunoreactivity and gene expression were coupled with larger increases of Dnm1 gene expression in 10-month-old glaucomatous ONH. Subcellular fractionation analysis indicates increased release of both OPA1 and cytochrome c from mitochondria in 10-month-old glaucomatous ONs. CONCLUSIONS: IOP elevation may directly damage mitochondria in the ONH axons by promoting reduction of COX, mitochondrial fission and cristae depletion, alterations of OPA1 and Dnm1 expression, and induction of OPA1 release. Thus, interventions to preserve mitochondria may be useful for protecting against ON degeneration in glaucoma.


Subject(s)
GTP Phosphohydrolases/biosynthesis , Gene Expression , Glaucoma/pathology , Intraocular Pressure/physiology , Mitochondria/ultrastructure , Optic Nerve/pathology , RNA, Messenger/genetics , Animals , Blotting, Western , Disease Models, Animal , Dynamin I/biosynthesis , Dynamin I/genetics , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/genetics , Female , GTP Phosphohydrolases/genetics , Glaucoma/metabolism , Glaucoma/physiopathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Optic Nerve/metabolism , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...