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.
Int J Mol Sci ; 23(8)2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35456925

ABSTRACT

Neuroinflammation is a crucial process for the loss of retinal ganglion cells (RGC), a major characteristic of glaucoma. High expression of high-mobility group box protein 1 (HMGB1) plays a detrimental role in inflammatory processes and is elevated in the retinas of glaucoma patients. Therefore, this study aimed to investigate the effects of the intravitreal injection of an anti-HMGB1 monoclonal antibody (anti-HMGB1 Ab) in an experimental animal model of glaucoma. Two groups of Spraque Dawley rats received episcleral vein occlusion to chronically elevate intraocular pressure (IOP): (1) the IgG group, intravitreal injection of an unspecific IgG as a control, n = 5, and (2) the HMGB1 group, intravitreal injection of an anti-HMGB1 Ab, n = 6. IOP, retinal nerve fiber layer thickness (RNFLT), and the retinal flash response were monitored longitudinally. Post-mortem examinations included immunohistochemistry, microarray, and mass spectrometric analysis. RNFLT was significantly increased in the HMGB1 group compared with the IgG group (p < 0.001). RGC density showed improved neuronal cell survival in the retina in HMGB1 compared with the IgG group (p < 0.01). Mass spectrometric proteomic analysis of retinal tissue showed an increased abundance of RNA metabolism-associated heterogeneous nuclear ribonucleoproteins (hnRNPs), such as hnRNP U, D, and H2, in animals injected with the anti-HMGB1 Ab, indicating that the application of the antibody may cause increased gene expression. Microarray analysis showed a significantly decreased expression of C-X-C motif chemokine ligand 8 (CXCL8, p < 0.05) and connective tissue growth factor (CTGF, p < 0.01) in the HMGB1 group. Thus, these data suggest that intravitreal injection of anti-HMGB1 Ab reduced HMGB1-dependent inflammatory signaling and mediated RGC neuroprotection.


Subject(s)
Glaucoma , HMGB1 Protein , Animals , Disease Models, Animal , Glaucoma/metabolism , Humans , Immunoglobulin G , Intraocular Pressure , Proteomics , Rats
2.
Exp Eye Res ; 213: 108853, 2021 12.
Article in English | MEDLINE | ID: mdl-34800481

ABSTRACT

PURPOSE: The roles of vascular dysfunction and chronic stress have been extensively discussed in the pathophysiology of glaucoma. Our aim was to test whether chronic stress causes retinal vascular dysfunction and therewith induces retinal ganglion cells (RGCs) loss. METHODS: Twelve mice underwent chronic social defeat (CSD) stress, while 12 mice received control treatment only. Intraocular pressure (IOP) was measured with a rebound tonometer. Blood plasma corticosterone concentration and adrenal gland weight were used to assess stress levels. Brn-3a staining in retinas and PPD staining in optic nerve cross sections were conducted to assess the survival of RGCs and axons respectively. The ET-1 and α-SMA levels were determined in retina. Retinal vascular autoregulation, functional response to various vasoactive agents and vascular mechanics were measured using video microscopy. RESULTS: No significant difference in IOP levels was observed during and after CSD between CSD mice and controls. CSD stress caused hypercortisolemia 2 days post-CSD. However, increased corticosterone levels went back to normal 8 months after CSD. CSD-exposed mice developed adrenal hyperplasia 3 days post-CSD, which was normalized by 8 months. RGC and axon survival were similar between CSD mice and controls. However, CSD stress caused irreversible, impaired autoregulation and vascular dysfunction of retinal arterioles in CSD mice. In addition, impaired maximal dilator capacity of retinal arterioles was observed 8 months post-CSD rather than 3 days post-CSD. Remarkably, ET-1 levels were increased 3 days post-CSD while α-SMA levels were decreased 8 months post-CSD. CONCLUSIONS: We found that CSD stress does not cause IOP elevation, nor loss of RGCs and their axons. However, it strikingly causes irreversible impaired autoregulation and endothelial function in murine retinal arterioles. In addition, CSD changed vascular mechanics on a long-term basis. Increased ET-1 levels and loss of pericytes in retina vessels may involve in this process.


Subject(s)
Retinal Artery/physiopathology , Retinal Diseases/physiopathology , Retinal Ganglion Cells/pathology , Social Defeat , Stress, Psychological/physiopathology , Actins/metabolism , Adrenal Hyperplasia, Congenital/physiopathology , Animals , Cell Survival , Chronic Disease , Corticosterone/blood , Disease Models, Animal , Disorder of Sex Development, 46,XY/physiopathology , Endothelin-1/metabolism , Intraocular Pressure/physiology , Male , Mice , Mice, Inbred C57BL , Ocular Hypertension/physiopathology , Optic Nerve/physiopathology , Retinal Artery/metabolism , Retinal Diseases/metabolism , Retinal Ganglion Cells/metabolism , Stress, Psychological/metabolism , Tonometry, Ocular , Transcription Factor Brn-3A/metabolism , Video Recording
3.
Brain Struct Funct ; 223(7): 3463-3471, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29936552

ABSTRACT

The antiphospholipid syndrome (APS) is an autoimmune disease characterized by the presence of antiphospholipid antibodies, which may trigger vascular thrombosis with consecutive infarcts. However, cognitive dysfunctions representing one of the most commonest neuropsychiatric symptoms are frequently present despite the absence of any ischemic brain lesions. Data on the structural and functional basis of the neuropsychiatric symptoms are sparse. To examine the effect of APS on hippocampal neurogenesis and on white matter, we induced experimental APS (eAPS) in adult female Balb/C mice by immunization with ß2-glycoprotein 1. To investigate cell proliferation in the dentate gyrus granular cell layer (DG GCL), eAPS and control mice (n = 5, each) were injected with 5-bromo-2'-deoxyuridine (BrdU) once a day for 10 subsequent days. Sixteen weeks after immunization, eAPS resulted in a significant reduction of BrdU-positive cells in the DG GCL compared to control animals. However, double staining with doublecortin and NeuN revealed a largely preserved neurogenesis. Ultrastructural analysis of corpus callosum (CC) axons in eAPS (n = 6) and control mice (n = 7) revealed no significant changes in CC axon diameter or g-ratio. In conclusion, decreased cellular proliferation in the hippocampus of eAPS mice indicates a limited regenerative potential and may represent one neuropathological substrate of cognitive changes in APS while evidence for alterations of white matter integrity is lacking.


Subject(s)
Antiphospholipid Syndrome/chemically induced , Antiphospholipid Syndrome/pathology , Cell Proliferation , Dentate Gyrus/pathology , Animals , Antibodies, Antiphospholipid/metabolism , Autoantigens/pharmacology , Behavior Rating Scale , Bromodeoxyuridine/administration & dosage , Bromodeoxyuridine/metabolism , Cell Differentiation/physiology , Corpus Callosum/ultrastructure , Disease Models, Animal , Female , Fluorescence , Mice , Mice, Inbred BALB C , Neurogenesis , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/metabolism , beta 2-Glycoprotein I/pharmacology
4.
PLoS One ; 12(4): e0175451, 2017.
Article in English | MEDLINE | ID: mdl-28384305

ABSTRACT

Purpose of this study was to investigate firstly specific proteomic changes within the retina in the course of an animal glaucoma model and to identify secondly new approaches for neuroprotective, therapeutic options in glaucoma by addressing those specific changes. Intraocular pressure was elevated through cauterization of episcleral veins in adult Sprague Dawley rats. Molecular and morphological changes were surveyed using mass spectrometry, optical coherence tomography as well as immunohistochemical cross section- and flat mount stainings. By quantifying more than 1500 retinal proteins, it was found that the HspB5 protein and numerous beta-crystallins showed a uniform and unique shifting expression pattern as a result of different periods of elevated IOP exposure. Crystallins showed a significant downregulation (p<0.05) after 3 weeks of elevated IOP and an upregulation after 7 weeks. Counteracting those typical changes, an intravitreal injection of ß-crystallin B2 at the time of IOP elevation was found to reduce retinal ganglion cell loss (p<0.05), decrease of the retinal nerve fiber layer (p<0.05) and impairment of the optic nerve. Ultimately, proteomic data revealed that ß-crystallin B2 might influence calcium-depended cell signaling pathways with severe effect on apoptosis and gene regulation. In this context especially annexin A5, calcium-transporting ATPase 1 and various histone proteins seem to play a major role.


Subject(s)
Disease Models, Animal , Glaucoma/pathology , Retinal Ganglion Cells/drug effects , beta-Crystallin B Chain/administration & dosage , Animals , Cell Survival/drug effects , Glaucoma/physiopathology , Intraocular Pressure , Intravitreal Injections , Male , Rats , Retinal Ganglion Cells/pathology , beta-Crystallin B Chain/pharmacology
5.
J Neuroinflammation ; 13(1): 82, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27090083

ABSTRACT

BACKGROUND: Elevated intraocular pressure (IOP), as well as fluctuations in IOP, is a main risk factor for glaucoma, but its pathogenic effect has not yet been clarified. Beyond the multifactorial pathology of the disease, autoimmune mechanisms seem to be linked to retinal ganglion cell (RGC) death. This study aimed to identify if intermittent IOP elevations in vivo (i) elicit neurodegeneration, (ii) provokes an immune response and (iii) whether progression of RGC loss can be attenuated by the B lymphocyte inhibitor Belimumab. METHODS: Using an intermittent ocular hypertension model (iOHT), Long Evans rats (n = 21) underwent 27 unilateral simulations of a fluctuating pressure profile. Nine of these animals received Belimumab, and additional seven rats served as normotensive controls. Axonal density was analyzed in PPD-stained optic nerve cross-sections. Retinal cross-sections were immunostained against Brn3a, Iba1, and IgG autoantibody depositions. Serum IgG concentration and IgG reactivities were determined using ELISA and protein microarrays. Data was analyzed using ANOVA and Tukey HSD test (unequal N) or student's independent t test by groups. RESULTS: A wavelike IOP profile led to a significant neurodegeneration of optic nerve axons (-10.6 %, p < 0.001) and RGC (-19.5 %, p = 0.02) in iOHT eyes compared with fellow eyes. Belimumab-treated animals only showed slightly higher axonal survival and reduced serum IgG concentration (-29 %) after iOHT. Neuroinflammatory events, indicated by significantly upregulated microglia activation and IgG autoantibody depositions, were shown in all injured retinas. Significantly elevated serum autoantibody immunoreactivities against glutathione-S-transferase, spectrin, and transferrin were observed after iOHT and were negatively correlated to the axon density. CONCLUSIONS: Intermittent IOP elevations are sufficient to provoke neurodegeneration in the optic nerve and the retina and elicit changes of IgG autoantibody reactivities. Although the inhibition of B lymphocyte activation failed to ameliorate axonal survival, the correlation between damage and changes in the autoantibody reactivity suggests that autoantibody profiling could be useful as a biomarker for glaucoma.


Subject(s)
Glaucoma/immunology , Nerve Degeneration/immunology , Ocular Hypertension/immunology , Animals , Autoantibodies/immunology , Disease Models, Animal , Glaucoma/pathology , Immunohistochemistry , Intraocular Pressure , Male , Nerve Degeneration/pathology , Ocular Hypertension/complications , Ocular Hypertension/pathology , Rats, Long-Evans , Retinal Ganglion Cells/pathology , Risk Factors
6.
Invest Ophthalmol Vis Sci ; 55(1): 600-11, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24408983

ABSTRACT

PURPOSE: An important, yet not exclusive, aspect of primary open angle glaucoma is elevated intraocular pressure (IOP) profiles within fluctuations and pressure peaks. The study aimed at establishing minimally invasive methods for recurrent IOP elevation in rats to investigate the impact of IOP dynamics and pathomorphologic retinal alterations during and after IOP elevation. METHODS: Intraocular pressure was elevated unilaterally in Long Evans rats to a level of ≈35 mm Hg for 1 hour in a total of 30 manipulations within 6 weeks, by using two methods: (1) suction-cup oculopression and (2) loop-adjusted oculopression. Retinal thickness (RT) was measured via optical coherence tomography (OCT), and neuronal survival was analyzed. Additional experiments were performed for "in situ" OCT investigations during exposures to different IOP levels. RESULTS: A mean IOP exposure of +737.3 ± 9.6 ΔIOP mm Hg for loop adjustment and +188.9 ± 16 ΔIOP mm Hg for suction cup was achieved. Optical coherence tomography examination revealed notable changes of RT between controls, untreated, and treated eyes, and evaluation of neuronal loss showed a significant decrease of retinal ganglion cell (RGC) density in both groups. In situ OCT investigation showed paradoxical retinal distortion and deformation of the optic nerve head toward the eye background. CONCLUSIONS: After accurate IOP elevation with minimally invasive methods, it was possible to detect RGC loss and retinal thinning. While suction cup is capable of simulating accurate arbitrary IOP profiles, loop adjustment enables the detection of pressure-dependent retinal alterations. For the first time, it was feasible to investigate consequences of variable IOP elevation profiles, including pressure peaks, by using real-time live imaging in vivo.


Subject(s)
Glaucoma, Open-Angle/physiopathology , Intraocular Pressure/physiology , Retinal Degeneration/diagnosis , Retinal Ganglion Cells/pathology , Tonometry, Ocular/methods , Animals , Disease Models, Animal , Disease Progression , Follow-Up Studies , Glaucoma, Open-Angle/complications , Glaucoma, Open-Angle/diagnosis , Male , Rats , Rats, Long-Evans , Retinal Degeneration/etiology , Retinal Degeneration/physiopathology , Tomography, Optical Coherence
7.
Mol Vis ; 19: 1804-14, 2013.
Article in English | MEDLINE | ID: mdl-23946635

ABSTRACT

PURPOSE: In recent years, numerous studies have investigated the involvement of immunological mechanisms in glaucoma. Until now, it has not been determined whether the altered antibody pattern detected in patients is harmful to retinal ganglion cells (RGCs) or triggers disease formation in any way. In a model of experimental autoimmune glaucoma, RGC loss can be induced through immunization with certain ocular antigens. In the current study, the time course of the levels of autoreactivity against ocular tissues after immunization was examined. METHODS: Intraocular pressure was measured regularly. Ten weeks after immunization with an optic nerve homogenate antigen (ONA), the number of RGCs was determined. Immunoglobulin G levels in aqueous humor were measured via enzyme-linked immunosorbent assay at the same time point. Serum from different time points was used to analyze the possible occurrence of autoreactive antibodies against the retina or optic nerve in this autoimmune glaucoma model. Additionally, optic nerve and brain sections were evaluated for possible pathological findings. RESULTS: Intraocular pressure stayed within the normal range throughout this study. A continuous increase of autoreactive antibodies against the optic nerve and retina sections was observed. At 4, 6, and 10 weeks, antibody reactivity was significantly higher in ONA animals (p<0.01). Aqueous humor immunoglobulin G levels were also significantly higher in the ONA group (p=0.006). Ten weeks after immunization, significantly fewer RGCs were noted in the ONA group (p=0.00003). The optic nerves from ONA animals exhibited damaged axons. No pathological findings appeared in any brain sections. CONCLUSIONS: Our findings suggest that these modified antibodies play a substantial role in mechanisms leading to RGC death. The slow dissolution of RGCs observed in animals with autoimmune glaucoma is comparable to the slow progressive RGC loss in glaucoma patients, thus making this a useful model to develop neuroprotective therapies in the future.


Subject(s)
Antigens/immunology , Autoimmune Diseases/immunology , Eye/immunology , Glaucoma/immunology , Immunity/immunology , Immunization , Optic Nerve/immunology , Animals , Aqueous Humor/metabolism , Autoantibodies/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/physiopathology , Axons/pathology , Brain/pathology , Cattle , Disease Models, Animal , Eye/pathology , Eye/physiopathology , Fundus Oculi , Glaucoma/pathology , Glaucoma/physiopathology , Immunoglobulin G/metabolism , Intraocular Pressure , Male , Optic Nerve/physiopathology , Rats , Rats, Inbred Lew , Retina/immunology , Retina/pathology , Retina/physiopathology , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/pathology
8.
PLoS One ; 7(7): e40616, 2012.
Article in English | MEDLINE | ID: mdl-22848388

ABSTRACT

BACKGROUND: Antibodies against retinal and optic nerve antigens are detectable in glaucoma patients. Recent studies using a model of experimental autoimmune glaucoma demonstrated that immunization with certain ocular antigens causes an immun-mediated retinal ganglion cell loss in rats. METHODOLOGY/PRINCIPAL FINDINGS: Rats immunized with a retinal ganglion cell layer homogenate (RGA) had a reduced retinal ganglion cell density on retinal flatmounts (p = 0.007) and a lower number of Brn3(+) retinal ganglion cells (p = 0.0001) after six weeks. The autoreactive antibody development against retina and optic nerve was examined throughout the study. The levels of autoreactive antibodies continuously increased up to 6 weeks (retina: p = 0.004; optic nerve: p = 0.000003). Additionally, antibody deposits were detected in the retina (p = 0.02). After 6 weeks a reactive gliosis (GFAP density: RGA: 174.7±41.9; CO: 137.6±36.8, p = 0.0006; %GFAP(+) area: RGA: 8.5±3.4; CO: 5.9±3.6, p = 0.006) as well as elevated level of Iba1(+) microglia cells (p = 0.003) was observed in retinas of RGA animals. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that these antibodies play a substantial role in mechanisms leading to retinal ganglion cell death. This seems to lead to glia cell activation as well as the invasion of microglia, which might be associated with debris clearance.


Subject(s)
Autoantibodies/immunology , Autoantigens/pharmacology , Glaucoma/immunology , Microglia/immunology , Retinal Ganglion Cells/immunology , Animals , Autoantigens/immunology , Cell Death/drug effects , Cell Death/immunology , Glaucoma/pathology , Immunization , Male , Microglia/pathology , Rats , Rats, Inbred Lew , Retinal Ganglion Cells/pathology
9.
J Neurol Sci ; 316(1-2): 108-11, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22326363

ABSTRACT

We report a novel heteroplasmic point mutation G8299A in the gene for mitochondrial tRNA(Lys) in a patient with progressive external ophthalmoplegia complicated by recurrent respiratory insufficiency. Biochemical analysis of respiratory chain complexes in muscle homogenate showed a combined complex I and IV deficiency. The transition does not represent a known neutral polymorphism and affects a position in the tRNA acceptor stem which is conserved in primates, leading to a destabilization of this functionally important domain. In vitro analysis of an essential maturation step of the tRNA transcript indicates the probable pathogenicity of this mutation. We hypothesize that there is a causal relationship between the novel G8299A transition and progressive external ophthalmoplegia with recurrent respiratory failure due to a depressed respiratory drive.


Subject(s)
Ophthalmoplegia, Chronic Progressive External/genetics , Point Mutation/genetics , RNA, Transfer, Lys/genetics , RNA/genetics , Respiratory Insufficiency/genetics , Base Sequence , Humans , Male , Middle Aged , Molecular Sequence Data , Ophthalmoplegia, Chronic Progressive External/complications , Ophthalmoplegia, Chronic Progressive External/diagnosis , RNA, Mitochondrial , Respiratory Insufficiency/complications , Respiratory Insufficiency/diagnosis , Secondary Prevention
SELECTION OF CITATIONS
SEARCH DETAIL
...