Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 61(3): 46, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32207812

ABSTRACT

Purpose: To scrutinize alterations in cellular interactions and cell signaling in the glaucomatous retina, mouse retinal explants were exposed to elevated pressure. Methods: Retinal explants were prepared from C57bl6 mice and cultivated in a pressure chamber under normotensive (atmospheric pressure + 0 mm Hg), moderately elevated (30 mm Hg), and highly elevated (60 mm Hg) pressure conditions. The expression levels of proteins involved in the formation of tight junctions (zonula occludens 1 [ZO-1], occludin, and claudin-5) and adherens junctions (VE-cadherin and ß-catenin) and in cell-signaling cascades (Cdc42 and activated Cdc42 kinase 1 [ACK1]), as well as the expression levels of the growth-factor receptors platelet-derived growth factor receptor beta and vascular endothelial growth factor receptors 1 and 2 (VEGFR-1, VEGFR-2) and of diverse intracellular proteins (ß-III-tubulin, glial fibrillary acidic protein transcript variant 1, α-smooth muscle actin, vimentin, and von Willebrand factor VIII), were analyzed using immunohistochemistry, western blotting, and quantitative real-time polymerase chain reactions. Results: The retinal explants were well preserved when cultured in the pressure chambers used in this study. The responses to pressure elevation varied among diverse retinal cells. Under elevated pressure, the expression of ZO-1 increased in the large vessels, neuronal cells began to express VEGFR-1, and the Cdc42 expression in the optic nerve head was downregulated. Overall we found significant transcriptional downregulation of VE-cadherin, ß-catenin, VEGFR-1, VEGFR-2, vimentin, Cdc42, and ACK1. Western blotting and immunohistochemistry indicated a loss of VE-cadherin with pressure elevation, whereas the protein levels of ZO-1, occludin, VEGFR-1, and ACK1 increased. Conclusions: The pressure chamber used for cultivating mouse retinal explants can serve as an in vitro model system for investigating molecular alterations in glaucoma. In this system, responses of the entire retinal cells toward elevated pressure with conspicuous changes in the vasculature and the optic nerve head can be seen. In particular, our investigations indicate that changes in the blood-retina barrier and in cellular signaling are induced by pressure elevation.


Subject(s)
Adherens Junctions/metabolism , Eye Proteins/metabolism , Glaucoma/metabolism , Retina/metabolism , Tight Junctions/metabolism , Animals , Antigens, CD/metabolism , Atmospheric Pressure , Blotting, Western , Cadherins/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Occludin/metabolism , Organ Culture Techniques , Protein-Tyrosine Kinases/metabolism , Real-Time Polymerase Chain Reaction , Zonula Occludens-1 Protein/metabolism , beta Catenin/metabolism , cdc42 GTP-Binding Protein/metabolism
2.
Exp Eye Res ; 185: 107676, 2019 08.
Article in English | MEDLINE | ID: mdl-31128101

ABSTRACT

Evidence of an age-related increase of ß-synuclein (SNCB) in several parts of the visual system including the retina has been reported. SNCB is thought to function as an antagonist of α-synuclein in neurodegenerative diseases, but the exact role of SNCB remains unclear. The presented work studies two different aspects of the onset and role of SNCB in the retinal pigment epithelium (RPE). First, the topographical and intracellular distributions of SNCB in the RPE of non-human marmoset monkey (Callithrix jacchus) were evaluated in paraffin-embedded eyes and RPE whole mounts from different developmental stages (neonatal, adolescent, and adult). Thus, revealed distinct lifetime-related alterations of the topographical and intracellular distributions of SNCB in the primate macula compared to the retinal periphery. Furthermore, the function and influences of SNCB on ARPE-19 cells and primary porcine RPE (ppRPE) cells were characterized by exposing these cells with recombinant SNCB (rSNCB) at different concentrations. Moreover, apoptosis, protein- and mRNA-expression levels of factors of the p53/MDM2 signaling cascade and inflammation- and oxidation-related genes were investigated. The observed dose-depended decreased apoptosis rates together with the PLD2 mediated activation of the p53 pathway promotes senescence-related processes in SNCB exposed common ARPE-19 cells from human origin. Further, increased HMOX1 and NOX4 levels indicate increased oxidative stress and inflammatory responses triggered by SNCB. The obtained differences in the distribution of SNCB in primate RPE together with alterations of cellular functions in rSNCB-exposed RPE cells (e.g., ARPE-19, ppRPE) support SNCB-related effects like inflammatory response and stress-related properties on RPE over lifetime. The possible functional relevance of SNCB in physiological aging converting into a pathophysiological condition should be investigated in further studies.


Subject(s)
Aging/physiology , Retina/metabolism , Retinal Pigment Epithelium/metabolism , beta-Synuclein/metabolism , Animals , Apoptosis , Callithrix , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/physiology , Heme Oxygenase (Decyclizing)/metabolism , Humans , Male , NADPH Oxidase 4/metabolism , Oxidative Stress , Paraffin Embedding , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retina/drug effects , Retina/pathology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Signal Transduction , Sus scrofa , Tumor Suppressor Protein p53/genetics , beta-Synuclein/pharmacology
3.
J Autoimmun ; 100: 75-83, 2019 06.
Article in English | MEDLINE | ID: mdl-30885419

ABSTRACT

Gene and protein expression profiles of iris biopsies, aqueous humor (AqH), and sera in patients with juvenile idiopathic arthritis-associated uveitis (JIAU) in comparison to control patients with primary open-angle glaucoma (POAG) and HLA-B27-positive acute anterior uveitis (AAU) were investigated. Via RNA Sequencing (RNA-Seq) and mass spectrometry-based protein expression analyses 136 genes and 56 proteins could be identified as being significantly differentially expressed (DE) between the JIAU and POAG group. Gene expression of different immunoglobulin (Ig) components as well as of the B cell-associated factors ID3, ID1, and EBF1 was significantly upregulated in the JIAU group as compared to POAG patients. qRT-PCR analysis showed a significantly higher gene expression of the B cell-related genes CD19, CD20, CD27, CD138, and MZB1 in the JIAU group. At the protein level, a significantly higher expression of Ig components in JIAU than in POAG was confirmed. The B cell-associated protein MZB1 showed a higher expression in JIAU patients than in POAG which was confirmed by western blot analysis. Using bead-based immunoassay analysis we were able to detect a significantly higher concentration of the B cell-activating and survival factors BAFF, APRIL, and IL-6 in the AqH of JIAU and AAU patients than in POAG patients. The intraocularly upregulated B cell-specific genes and proteins in iris tissue suggest that B cells participate in the immunopathology of JIAU. The intracameral environment in JIAU may facilitate local effector and survival functions of B cells, leading to disease course typical for anterior uveitis.


Subject(s)
Aqueous Humor/immunology , Arthritis, Juvenile/immunology , Eye Proteins/immunology , Gene Expression Regulation/immunology , Iris/immunology , Transcriptome/immunology , Uveitis/immunology , Adolescent , Adult , Aged , Arthritis, Juvenile/complications , Arthritis, Juvenile/pathology , Child , Child, Preschool , Female , Humans , Iris/pathology , Male , Middle Aged , Proteomics , Uveitis/etiology , Uveitis/pathology
4.
Cell Transplant ; 27(5): 796-813, 2018 05.
Article in English | MEDLINE | ID: mdl-29808713

ABSTRACT

Increased ß-synuclein (Sncb) expression has been described in the aging visual system. Sncb functions as the physiological antagonist of α-synuclein (Snca), which is involved in the development of neurodegenerative diseases, such as Parkinson's and Alzheimer's diseases. However, the exact function of Sncb remains unknown. The aim of this study was to elucidate the age-dependent role of Sncb in brain microvascular endothelial cells (BMECs). BMECs were isolated from the cortices of 5- to 9-d-old Sprague-Dawley rats and were cultured with different concentrations of recombinant Sncb (rSncb) up to 72 h resembling to some degree age-related as well as pathophysiological conditions. Viability, apoptosis, expression levels of Snca, and the members of phospholipase D2 (Pld2)/ p53/ Mouse double minute 2 homolog (Mdm2)/p19(Arf) pathway, response in RAC-alpha serine/threonine-protein kinase (Akt), and stress-mediating factors such as heme oxygenase (decycling) 1 (Hmox) and Nicotinamide adenine dinucleotide phosphate oxygenase 4 (Nox4) were examined. rSncb-induced effects were confirmed through Sncb small interfering RNA (siRNA) knockdown in BMECs. We demonstrated that the viability decreases, while the rate of apoptosis underly dose-dependent alterations. For example, apoptosis increases in BMECs following the treatment with higher dosed rSncb. Furthermore, we observed a decrease in Snca immunostaining and messenger RNA (mRNA) levels following the exposure to higher rScnb concentrations. Akt was shown to be downregulated and pAkt upregulated by this treatment, which was accompanied by a dose-independent increase in p19(Arf) levels and enhanced intracellular Mdm2 translocation in contrast to a dose-dependent p53 activation. Moreover, Pld2 activity was shown to be induced in rSncb-treated BMECs. The expression of Hmox and Nox4 after Sncb treatment was altered on BEMCs. The obtained results demonstrate dose-dependent effects of Sncb on BMECs in vitro. For example, the p53-mediated and Akt-independent apoptosis together with the stress-mediated response of BMECs related to exposure of higher SNCB concentrations may reflect the increase in Sncb with duration of culture as well as its impact on cell decay. Further studies, expanding on the role of Sncb, may help understand its role in the neurodegenerative diseases.


Subject(s)
Aging/metabolism , Apoptosis , Brain/blood supply , Endothelial Cells/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , beta-Synuclein/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Female , Heme Oxygenase-1/metabolism , Microvessels/pathology , NADPH Oxidase 4/metabolism , Phospholipase D/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , alpha-Synuclein/metabolism
5.
Restor Neurol Neurosci ; 36(1): 31-43, 2018.
Article in English | MEDLINE | ID: mdl-29439367

ABSTRACT

BACKGROUND: Retinal ganglion cells (RGCs) of mammals lose the ability to regenerate injured axons during postnatal maturation, but little is known about the underlying molecular mechanisms. OBJECTIVE: It remains of particular importance to understand the mechanisms of axonal regeneration to develop new therapeutic approaches for nerve injuries. METHODS: Retinas from newborn to adult monkeys (Callithrix jacchus)1 were obtained immediately after death and cultured in vitro. Growths of axons were monitored using microscopy and time-lapse video cinematography. Immunohistochemistry, Western blotting, qRT-PCR, and genomics were performed to characterize molecules associated with axonal regeneration and growth. A genomic screen was performed by using retinal explants versus native and non-regenerative explants obtained from eye cadavers on the day of birth, and hybridizing the mRNA with cross-reacting cDNA on conventional human microarrays. Followed the genomic screen, siRNA experiments were conducted to identify the functional involvement of identified candidates. RESULTS: Neuron-specific human ribonucleoprotein N (snRPN) was found to be a potential regulator of impaired axonal regeneration during neuronal maturation in these animals. In particular, up-regulation of snRPN was observed during retinal maturation, coinciding with a decline in regenerative ability. Axon regeneration was reactivated in snRPN-knockout retinal ex vivo explants of adult monkey. CONCLUSION: These results suggest that coordinated snRPN-driven activities within the neuron-specific ribonucleoprotein complex regulate the regenerative ability of RGCs in primates, thereby highlighting a potential new role for snRPN within neurons and the possibility of novel postinjury therapies.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Retina/cytology , Retinal Ganglion Cells/cytology , snRNP Core Proteins/physiology , Age Factors , Animals , Animals, Newborn , Callithrix , Cells, Cultured , In Vitro Techniques , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Time Factors , Up-Regulation/genetics , snRNP Core Proteins/genetics
6.
Klin Monbl Augenheilkd ; 234(10): 1266-1275, 2017 Oct.
Article in German | MEDLINE | ID: mdl-28470655

ABSTRACT

Background In the early diagnosis of clinical glaucoma, peripapillary bleedings were almost pathognomonic for a capillary insult. In the perfusion diagnostics, it is predominantly accepted that perfusion imbalances and IOP-induced changes occur and play a crucial role. Biomechanical peculiarities of the optic nerve head and cellular responses to astrocytes are also likely involved. Material and Methods We present in vivo and ex vivo models of IOP-elevation to enhance the resolution of examining cellular and molecular changes and to understand the mechanisms of capillary changes due to IOP-elevation. Results The in vivo model consists of cauterization-caused elevation of IOP in rat eyes. Two to 3 veins were cauterized to increase outflow resistance. The retinas were analyzed several weeks later and we found an abnormal expression of the neuron-specific molecule beta-III-tubulin in the capillary endothelium cells and in the vascular pericytes. Combined immunohistochemical stainings with different markers for various retinal cells confirmed the findings. The isolation of capillary endothelium cells and pericytes from rat brains (BMECs) and retinas (RMECs), and their cultivation under elevated IOP in vitro, confirmed the in vivo results. Conclusion The unexpected capillary response to elevation of IOP in vivo and in vitro could be seen as an early response of cells with expression of abnormal proteins. This result may explain clinical observations which dominate as peripapillary bleedings or microinfarctions and are likely associated with the glaucoma-induced opticopathy.


Subject(s)
Glaucoma , Intraocular Pressure , Retinal Ganglion Cells , Animals , Capillaries , Disease Models, Animal , Glaucoma/physiopathology , Rats , Retina , Retinal Ganglion Cells/physiology
7.
Invest Ophthalmol Vis Sci ; 57(11): 4536-52, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27583825

ABSTRACT

Purpose: The primate central retina is characterized by an avascular fovea and well-defined perifoveal capillary plexus. Neither blood vessels nor their accompanying astrocytes enter the fovea during any stage of retinal development; a balance of angiogenic and angiostatic factors probably maintains foveal avascularity throughout life. The aim of this study was to identify potentially angiorepulsive factors involved in the development of the avascular primate retinal fovea. Methods: Retinas of newborn, juvenile, and adult Callithrix jacchus and Macaca fascicularis monkeys and control human retinas were studied to determine the localization of angiostatin relative to III ß-tubulin, glial fibrillary acidic protein, vascular endothelial growth factor (VEGF), platelet endothelial cell adhesion molecule-1 (PECAM), and the angiostatin receptor αvß3-integrin in the foveal, macular, and peripheral retina. Expression studies were performed using immunohistochemistry (IHC) on retinal whole-mount and paraffin sections, and Western blotting on frozen material. The complex network of the main retinal cell types was identified by IHC of retinal whole mounts. Results: In general, lifetime expression of angiostatin was found in all retinas. Colabeling with different markers revealed retinal ganglion cells as the main source of angiostatin expression in the primate retina, whereas PECAM-immunopositive blood capillaries expressed the angiostatin receptor αvß3-integrin, and capillary-associated astrocytes expressed VEGF. Conclusions: This study provides the first evidence of angiostatin expression in the primate retina; the expression of angiostatin in the avascular foveal region and the peripheral retina suggests that angiostatin may play a role in the regulation of retinal vascularization, providing a possible explanation for the development and persistence of an avascular fovea.


Subject(s)
Angiostatins/biosynthesis , Fovea Centralis/blood supply , Retinal Vessels/metabolism , Aged , Animals , Animals, Newborn , Blotting, Western , Callithrix , Capillaries/cytology , Capillaries/metabolism , Female , Fovea Centralis/cytology , Fovea Centralis/metabolism , Glial Fibrillary Acidic Protein/biosynthesis , Humans , Immunohistochemistry , Macaca fascicularis , Male , Middle Aged , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism
8.
Lab Invest ; 96(4): 409-27, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26779828

ABSTRACT

To determine the role of high-mobility group box 1 protein (HMGB-1) in cellular and tissue models of elevated pressure-induced neurodegeneration, regeneration, and inflammation. Mouse retinal photoreceptor-derived cells (661W) and retinal explants were incubated either under elevated pressure or in the presence of recombinant HMGB-1 (rHMGB-1) to investigate the mechanisms of response of photoreceptors. Immunohistochemistry, western blotting, and the quantitative real-time PCR were used to examine the expression levels of immunological factors (eg, HMGB-1, receptor for advanced glycation end products (RAGE)), Toll-like receptors 2 and 4 (TLR-2, TLR-4), apoptosis-related factors (eg, B-cell lymphoma 2 (Bcl-2), Bcl-2-associated death promoter (Bad)) as well as cytokine expression (eg, tumor necrosis factor alpha (TNF-α), interleukin (IL)-4, IL-6, and vascular endothelial growth factor (VEGF)). The data revealed increased the expression of HMGB-1 and its receptors RAGE, TLR-2, and TLR-4, and TNF-α as well as pro-apoptotic factors (eg, Bad) as well as apoptosis in 661W cells exposed to elevated pressure. Co-cultivation of 661W cells with rHMGB-1 increased the expression levels of pro-apoptotic Bad and cleaved Caspase-3 resulting in apoptosis. Cytokine array studies revealed an increased release of TNF-α, IL-4, IL-6, and VEGF after incubation of 661W cells with rHMGB-1. Upregulation of HMGB-1, TLR-2, and RAGE as well as anti-apoptotic Bcl-2 expression levels was found in the retinal explants exposed to rHMGB-1 or elevated pressure. The results suggest that HMGB-1 promotes an inflammatory response and mediates apoptosis in the pathology of photoreceptors and retinal homeostasis. HMGB-1 may have a key role in ongoing damage of retinal cells under conditions of elevated intraocular pressure.


Subject(s)
Apoptosis/drug effects , HMGB1 Protein/pharmacology , Recombinant Proteins/pharmacology , Retina/drug effects , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Line , Cytokines/metabolism , Gene Expression/drug effects , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Male , Mice, Inbred C57BL , Microscopy, Fluorescence , Pressure , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Retina/cytology , Retina/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
9.
Front Neuroanat ; 9: 16, 2015.
Article in English | MEDLINE | ID: mdl-25788877

ABSTRACT

Four distinct proteins are regulated in the aging neuroretina and may be regulated in the cerebral cortex, too: peroxiredoxin, beta-synuclein, PARK[Parkinson disease(autosomal recessive, early onset)]7/DJ-1, and Stathmin. Thus, we performed a comparative analysis of these proteins in the the primary somatosensory cortex (S1) and primary visual cortex (V1) in rats, in order to detect putative common development-, maturation- and age-related changes. The expressions of peroxiredoxin, beta-synuclein, PARK[Parkinson disease (autosomal recessive, early onset)]7/DJ-1, and Stathmin were compared in the newborn, juvenile, adult, and aged S1 and V1. Western blot (WB), quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and immunohistochemistry (IHC) analyses were employed to determine whether the changes identified by proteomics were verifiable at the cellular and molecular levels. All of the proteins were detected in both of the investigated cortical areas. Changes in the expressions of the four proteins were found throughout the life-time of the rats. Peroxiredoxin expression remained unchanged over life-time. Beta-Synuclein expression was massively increased up to the adult stage of life in both the S1 and V1. PARK[Parkinson disease (autosomal recessive, early onset)]7/DJ-1 exhibited a massive up-regulation in both the S1 and V1 at all ages. Stathmin expression was massively down regulated after the neonatal period in both the S1 and V1. The detected protein alterations were analogous to their retinal profiles. This study is the first to provide evidence that peroxiredoxin, beta-synuclein, PARK[Parkinson disease (autosomal recessive, early onset)]7/DJ-1, and Stathmin are associated with postnatal maturation and aging in both the S1 and V1 of rats. These changes may indicate their involvement in key functional pathways and may account for the onset or progression of age-related pathologies.

10.
Cell Transplant ; 24(9): 1829-44, 2015.
Article in English | MEDLINE | ID: mdl-25299378

ABSTRACT

The purpose of the study was to further scrutinize the potential of ßB2-crystallin in supporting regeneration of injured retinal ganglion cell axons both in vitro and in vivo. Retinal explants obtained from animals after treatment either with lens injury (LI) alone or with combined LI 5 days or 3 days before or simultaneously with an optic nerve crush (ONC) were cultured for 96 h under regenerative conditions, and the regenerating axons were quantified and compared with untreated controls. These measurements were then repeated with LI replaced by intravitreal injections of γ-crystallin and ß-crystallin at 5 days before ONC. Finally, ßB2-crystallin-overexpressing transfected neural progenitor cells (ßB2-crystallin-NPCs) in the eye were studied after crushing the optic nerve in vivo. Regeneration was monitored with the aid of immunoblotting of the retina and optic nerve both distal and proximal to the lesion site, and this was compared with controls that received injections of phosphate buffer only. LI performed 5 days or 3 days before ONC significantly promoted axonal outgrowth in vitro (p < 0.001), while LI performed alone before explantation did not. Intravitreal injections of ß-crystallin and γ-crystallin mimicked the effects of LI and significantly increased axonal regeneration in culture at the same time intervals (p < 0.001). Western blot analysis revealed that crystallins were present in the proximal optic nerve stump at the lesion site in ONC, but were neither expressed in the undamaged distal optic nerve nor in uninjured tissue. ßB2-crystallin-NPCs supported the regeneration of cut optic nerve axons within the distal optic nerve stump in vivo. The reported data suggest that ßB2-crystallin-producing "cell factories" could be used to provide novel therapeutic drugs for central nervous system injuries.


Subject(s)
Optic Nerve Injuries/therapy , Optic Nerve/pathology , Retinal Ganglion Cells/transplantation , beta-Crystallin B Chain/metabolism , Animals , Axons/physiology , Cells, Cultured , Immunohistochemistry , Injections, Intravenous , Intravitreal Injections , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Optic Nerve/metabolism , Rats , Rats, Sprague-Dawley , Regeneration , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/cytology , beta-Crystallin B Chain/administration & dosage , beta-Crystallin B Chain/genetics , gamma-Crystallins/administration & dosage , gamma-Crystallins/genetics , gamma-Crystallins/metabolism
11.
Graefes Arch Clin Exp Ophthalmol ; 251(1): 63-79, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23073841

ABSTRACT

BACKGROUND: Crystallins are thought to play a cytoprotective role in conditions of cellular stress. The aim of this study was to determine the effects of crystallin-ß-b2 (cryß-b2) and crystallin-ß-b3 (cryß-b3) on ARPE-19 cells in vitro and on the retinal pigment epithelium (RPE) in vivo. METHODS: The influence of cryß-b2 and cryß-b3 on the viability, proliferation and dying of ARPE-19 was measured by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium assay, bromo-2-deoxyuridine assay and life/death assay. The expressions of cryß-b2, cryß-b3, glial-derived neurotrophic factor (GDNF), and galectin-3 (Gal-3) in ARPE-19 cells were evaluated using immunohistochemistry (IHC), Western blotting (WB) and real-time-quantitative-PCR (qRT-PCR). To evaluate the response of cryß-b2 and cryß-b3 to stressed ARPE-19 cells, the cells were exposed to UV-light. In a rat model, cryß-b2-expressing neural progenitor cells (cryß-b2-NPCs) were injected intravitreally after retinal stress induced by optic nerve axotomy to examine whether they influence the RPE. Protein expression was examined 2 and 4 weeks postsurgery using IHC and WB. RESULTS: Detectable alterations of GDNF, and Gal-3 were found in ARPE-19 cells upon exposure to UV light. Adding the crystallins to the medium promoted proliferation and increased viability of ARPE-19 cells in vitro. The obtained data support the view that these crystallins possess epithelioprotective properties. Likewise, in vivo, intravitreally injected cryß-b2 and transplanted cryß-b2-NPCs protected RPE from indirectly induced stress. CONCLUSIONS: The data suggest that the RPE response to retinal ganglion cell denegeration is mediated via crystallins, which may thus be used therapeutically.


Subject(s)
Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , beta-Crystallin B Chain/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cell Survival , Cells, Cultured , Embryonic Stem Cells , Female , Galectin 3/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Immunohistochemistry , Intravitreal Injections , Male , Nerve Crush , Neural Stem Cells , Pregnancy , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/radiation effects , Transfection , Ultraviolet Rays
12.
Invest Ophthalmol Vis Sci ; 53(13): 8265-79, 2012 Dec 17.
Article in English | MEDLINE | ID: mdl-23132806

ABSTRACT

PURPOSE: Crystallin ß-b2 (crybb2) is known to support the regeneration of retinal ganglion cell (RGC) axons in culture. We investigated whether neuronal progenitor cells (NPCs) overexpressing crybb2 (crybb2-NPC) affect secondary retinal degeneration due to optic nerve crush in vivo. METHODS: NPCS were produced by dissociation and propagation of rat embryonic neural tube and eye primordial cells at embryonic days 13.5 and 15. Retinal degeneration was induced by injured optic nerve crush (BY suture, 20 seconds). Several groups were built: crybb2-NPC were injected into the vitreous body, while the Controls were comprised of recombinant crybb2-injected and PBS-injected groups. The eyes, in particular the retina, were analyzed by immunohistochemistry and Western blotting for different antigens at 2 and 4 weeks after surgery. RESULTS: At 2 and 4 weeks post surgery, crybb2-NPC resided within the vitreoretinal compartment, and were persistently nestin-positive throughout the experimental period. The cells stained positive for various neurotrophins and acted as "living" cell factories to support the survival of injured RGCs. The crybb2-NPC migrated throughout the eye structures and sometimes became integrated within the tissue. Most of the ocular cells responded to the appearance of crybb2-NPC with marked changes of certain proteins, including Iba-1 (microglia), vimentin (glial cells), and rhodopsin (photoreceptors). Photoreceptors also displayed a better survival after crybb2-NPC injection compared to control groups. CONCLUSIONS: Crybb2-NPC exert beneficial effects on the vitreoretinal compartment, which suggests that modified crybb2-NPC could be used in a novel strategy for the treatment of degenerative vitreoretinal diseases. However, future studies must determine the safety of in vivo administration of crybb2-NPC.


Subject(s)
Disease Models, Animal , Gene Expression Regulation/physiology , Neural Stem Cells/physiology , Photoreceptor Cells, Vertebrate/cytology , Retinal Degeneration/prevention & control , Retinal Ganglion Cells/cytology , beta-Crystallin B Chain/genetics , Animals , Biomarkers/metabolism , Blotting, Western , Cell Movement/physiology , Cell Survival/physiology , Female , Immunohistochemistry , Intravitreal Injections , Male , Nerve Crush , Nerve Growth Factors/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Stem Cell Transplantation , Transfection
13.
Exp Eye Res ; 92(2): 128-37, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21147101

ABSTRACT

To examine retinal angiogenesis in the Royal College of Surgeons rat (RCS) serving as a model for ischemic proliferative retinopathies at morphological, proteomic and mRNA levels in order to evaluate the interplay of morphological and molecular changes in the course of the disease. Photoreceptor degeneration was confirmed by histological cross-sections and optical coherence tomography. The capillary retinal network was visualized in RCS rats aged between 14 and 45 days (P14-P45) by perfusion with high molecular weight fluorescein isothiocyanate-labeled dextran and compared with corresponding Sprague-Dawley rats. Vascular crosstalks within central areas to peripheral retinal eccentricities were analyzed. The expression of vascular growth-associated factors and their receptors in the course of the abnormal vascular development, namely vascular endothelial growth factor (VEGF), VEGF receptor subtype 1 (VEGF-R1) and -2 (VEGF-R2), somatostatin (SRIF), somatostatin receptor subtype 2 (Sstr-2), angiopoietin-2 (Ang-2) and tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (Tie-2), was analyzed by immunohistochemistry, western blotting and quantitative real-time polymerase chain reaction. Underperfused areas without capillarization were found in the middle and peripheral retinal eccentricities of RCS rats until P29. Through the course of vascularization previously low perfused areas became completely perfused, but were characterized by significantly increased neovascularizations. Western blotting revealed specific expression of growth-associated factors and their receptors in the course of capillary development. VEGF was significantly increased until P29 in RCS rats, while SRIF was significantly upregulated at P21 and P29 at proteomic level in SD rats. At mRNA level Ang-2 was significantly upregulated in RCS rats at P29, VEGF-R1 and VEGF-R2 at P36 and SRIF at P36. Initial incomplete perfusion is followed by aberrant vessel formation. VEGF, SRIF, Ang-2 and their receptors are regulated at protein and mRNA levels, providing therapeutic possibilities for treating ischemic proliferative retinopathies in the course of the disease.


Subject(s)
Angiopoietin-2/metabolism , Retinal Degeneration/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Somatostatin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-2/genetics , Animals , Blotting, Western , Capillaries/metabolism , Capillaries/pathology , Dextrans , Disease Models, Animal , Fluorescein Angiography , Fluoresceins , Fluorescent Antibody Technique, Indirect , Microscopy, Fluorescence , RNA, Messenger/genetics , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Receptors, Somatostatin/metabolism , Retinal Degeneration/pathology , Retinal Neovascularization/pathology , Retinal Vessels/pathology , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
J Immunol ; 175(9): 5975-80, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16237091

ABSTRACT

Systemic lupus erythematosus T cells display decreased amounts of TCR zeta mRNA that results in part from limited binding of the transcriptional enhancer Elf-1 to the TCR zeta promoter. We have identified a new cis-binding site for the cAMP response element (CRE) modulator (CREM) on the TCR zeta promoter, centered on the -390 nucleotide. Transfection of T cells with an antisense CREM alpha plasmid reduced the binding of CREM to the TCR zeta promoter, as shown by chromatin and reporter chromatin immunoprecipitation assays, and enhanced the production of TCR zeta mRNA and protein. Mutagenesis of the -390 CRE site prevented the binding of CREM to the TCR zeta promoter. The mechanism of CREM-mediated repression appears to be chromatin dependent, because antisense CREM promotes the acetylation of histones on the TCR zeta promoter. Finally, we established an enhanced binding of CREM to the TCR zeta-chain promoter in systemic lupus erythematosus cells compared with control T cells. Our studies demonstrate that CREM alpha binds to the TCR zeta promoter and repress its activity.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Gene Expression Regulation , Membrane Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Chromatin Assembly and Disassembly , DNA-Binding Proteins/physiology , Humans , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Promoter Regions, Genetic , T-Lymphocytes/metabolism , Transcription Factors/physiology , Transcription, Genetic
15.
J Biol Chem ; 277(44): 41879-87, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12167632

ABSTRACT

S100A9, also referred to as MRP14, is a calcium-binding protein whose expression is tightly regulated during differentiation of myeloid cells. The present study was performed to study the cell type- and differentiation-specific transcriptional regulation of the S100A9 gene. Analysis of the S100A9 promoter in MonoMac-6 cells revealed evidence for a novel regulatory region from position -400 to -374 bp, termed myeloid-related protein regulatory element (MRE). MRE deletion resulted in a 5.2-fold reduction of promoter activity. By electrophoretic mobility shift analysis two nuclear complexes binding to this region were identified and referred to as MRE-binding complex A (MbcA) and MRE-binding complex B (MbcB). By mutagenesis the MRE-binding motif could be narrowed to a 12-bp region. The relevance of MRE is deduced from the observations that the formation of either MRE-binding complex A or MRE-binding complex B strongly correlated with S100A9 gene expression in a cell type-specific, activation- and differentiation-dependent manner. Moreover, DNA affinity chromatography and Western blot studies indicate that a Kruppel-related zinc finger protein and the transcriptional intermediary factor 1beta (TIF1beta) are involved in an MRE-binding complex, thereby regulating the S100A9 gene expression.


Subject(s)
Calgranulin B/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, Regulator/physiology , Promoter Regions, Genetic , Base Sequence , Bone Marrow/metabolism , Cell Differentiation , Cell Line , DNA-Binding Proteins/physiology , Humans , Molecular Sequence Data , Protein Binding , Repressor Proteins/physiology , Tripartite Motif-Containing Protein 28 , Zinc Fingers/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...