Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 21(1): 56, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38388518

ABSTRACT

Inherited, age-related, and acute retinal diseases are often exacerbated by an aberrant or excessive activity of the complement system. Consequently, cells not directly affected by an acute event or genetic variants may degenerate, resulting in enhanced visual impairment. The therapeutic potential of supplementation of complement factor H (FH), a key regulator of the complement cascade, is therefore particularly promising in the context of retinal diseases caused by complement activation. In this study, we engineered adeno-associated viruses (AAVs) containing sequences of two truncated human FH variants. The expression of these variants was regulated by the glial fibrillary acidic protein (GFAP) promoter, which is selectively active in gliotic Müller cells. Both FH variants consisted of FH domains 19-20, which were connected to domains 1-4 and 1-7, respectively, by a polyglycine linker. These AAVs were intravitreally injected following ischemic injury of C57BL/6J mouse retinas. We observed transgene expression in gliotic Müller cells and to some extent in astrocytes. The expression correlated directly with damage severity. Interventions resulted in decreased complement activation, accelerated normalization of microglia activity and morphological improvements. Reduced levels of C3 transcripts and C3d protein in conjunction with higher transcript levels of inhibitory regulators like Cfi and Cfh, hinted at attenuated complement activity. This study demonstrates the great potential of complement regulatory gene addition therapy. With further in vivo testing it could be applied to treat a wide range of retinal diseases where no causative therapies are available.


Subject(s)
Gliosis , Retinal Diseases , Mice , Animals , Humans , Gliosis/metabolism , Complement Factor H/genetics , Mice, Inbred C57BL , Retina/metabolism
2.
J Neuroinflammation ; 21(1): 33, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38273366

ABSTRACT

Diabetic retinopathy (DR) is considered a primarily microvascular complication of diabetes. Müller glia cells are at the centre of the retinal neurovascular unit and play a critical role in DR. We therefore investigated Müller cell-specific signalling pathways that are altered in DR to identify novel targets for gene therapy. Using a multi-omics approach on purified Müller cells from diabetic db/db mice, we found the mRNA and protein expression of the glucocorticoid receptor (GR) to be significantly decreased, while its target gene cluster was down-regulated. Further, oPOSSUM TF analysis and ATAC- sequencing identified the GR as a master regulator of Müller cell response to diabetic conditions. Cortisol not only increased GR phosphorylation. It also induced changes in the expression of known GR target genes in retinal explants. Finally, retinal functionality was improved by AAV-mediated overexpression of GR in Müller cells. Our study demonstrates an important role of the glial GR in DR and implies that therapeutic approaches targeting this signalling pathway should be aimed at increasing GR expression rather than the addition of more ligand.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Animals , Mice , Diabetes Mellitus/metabolism , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Ependymoglial Cells/metabolism , Neuroglia/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Retina/metabolism
3.
Glia ; 71(2): 391-414, 2023 02.
Article in English | MEDLINE | ID: mdl-36334068

ABSTRACT

The human macula is a highly specialized retinal region with pit-like morphology and rich in cones. How Müller cells, the principal glial cell type in the retina, are adapted to this environment is still poorly understood. We compared proteomic data from cone- and rod-rich retinae from human and mice and identified different expression profiles of cone- and rod-associated Müller cells that converged on pathways representing extracellular matrix and cell adhesion. In particular, epiplakin (EPPK1), which is thought to play a role in intermediate filament organization, was highly expressed in macular Müller cells. Furthermore, EPPK1 knockout in a human Müller cell-derived cell line led to a decrease in traction forces as well as to changes in cell size, shape, and filopodia characteristics. We here identified EPPK1 as a central molecular player in the region-specific architecture of the human retina, which likely enables specific functions under the immense mechanical loads in vivo.


Subject(s)
Ependymoglial Cells , Proteome , Humans , Mice , Animals , Proteome/metabolism , Proteomics , Retina/metabolism , Retinal Cone Photoreceptor Cells , Neuroglia/metabolism
4.
Hum Mol Genet ; 32(3): 431-449, 2023 01 13.
Article in English | MEDLINE | ID: mdl-35997788

ABSTRACT

Usher syndrome (USH) is the most common form of hereditary deaf-blindness in humans. USH is a complex genetic disorder, assigned to three clinical subtypes differing in onset, course and severity, with USH1 being the most severe. Rodent USH1 models do not reflect the ocular phenotype observed in human patients to date; hence, little is known about the pathophysiology of USH1 in the human eye. One of the USH1 genes, USH1C, exhibits extensive alternative splicing and encodes numerous harmonin protein isoforms that function as scaffolds for organizing the USH interactome. RNA-seq analysis of human retinae uncovered harmonin_a1 as the most abundant transcript of USH1C. Bulk RNA-seq analysis and immunoblotting showed abundant expression of harmonin in Müller glia cells (MGCs) and retinal neurons. Furthermore, harmonin was localized in the terminal endfeet and apical microvilli of MGCs, presynaptic region (pedicle) of cones and outer segments (OS) of rods as well as at adhesive junctions between MGCs and photoreceptor cells (PRCs) in the outer limiting membrane (OLM). Our data provide evidence for the interaction of harmonin with OLM molecules in PRCs and MGCs and rhodopsin in PRCs. Subcellular expression and colocalization of harmonin correlate with the clinical phenotype observed in USH1C patients. We also demonstrate that primary cilia defects in USH1C patient-derived fibroblasts could be reverted by the delivery of harmonin_a1 transcript isoform. Our studies thus provide novel insights into PRC cell biology, USH1C pathophysiology and development of gene therapy treatment(s).


Subject(s)
Usher Syndromes , Humans , Usher Syndromes/genetics , Usher Syndromes/therapy , Usher Syndromes/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Retina/metabolism , Photoreceptor Cells/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
5.
J Extracell Vesicles ; 11(9): e12254, 2022 09.
Article in English | MEDLINE | ID: mdl-36043482

ABSTRACT

Cell-cell interactions in the central nervous system are based on the release of molecules mediating signal exchange and providing structural and trophic support through vesicular exocytosis and the formation of extracellular vesicles. The specific mechanisms employed by each cell type in the brain are incompletely understood. Here, we explored the means of communication used by Müller cells, a type of radial glial cells in the retina, which forms part of the central nervous system. Using immunohistochemical, electron microscopic, and molecular analyses, we provide evidence for the release of distinct extracellular vesicles from endfeet and microvilli of retinal Müller cells in adult mice in vivo. We identify VAMP5 as a Müller cell-specific SNARE component that is part of extracellular vesicles and responsive to ischemia, and we reveal differences between the secretomes of immunoaffinity-purified Müller cells and neurons in vitro. Our findings suggest extracellular vesicle-based communication as an important mediator of cellular interactions in the retina.


Subject(s)
Extracellular Vesicles , Neuroglia , Animals , Ependymoglial Cells/metabolism , Mice , Neuroglia/metabolism , Neurons/metabolism , Retina/metabolism
6.
Front Immunol ; 13: 895519, 2022.
Article in English | MEDLINE | ID: mdl-35784369

ABSTRACT

The cellular events that dictate the initiation of the complement pathway in ocular degeneration, such as age-related macular degeneration (AMD), is poorly understood. Using gene expression analysis (single cell and bulk), mass spectrometry, and immunohistochemistry, we dissected the role of multiple retinal and choroidal cell types in determining the complement homeostasis. Our scRNA-seq data show that the cellular response to early AMD is more robust in the choroid, particularly in fibroblasts, pericytes and endothelial cells. In late AMD, complement changes were more prominent in the retina especially with the expression of the classical pathway initiators. Notably, we found a spatial preference for these differences. Overall, this study provides insights into the heterogeneity of cellular responses for complement expression and the cooperation of neighboring cells to complete the pathway in healthy and AMD eyes. Further, our findings provide new cellular targets for therapies directed at complement.


Subject(s)
Endothelial Cells , Macular Degeneration , Choroid , Complement System Proteins , Humans , Macular Degeneration/genetics , Retina
7.
Front Pharmacol ; 12: 771571, 2021.
Article in English | MEDLINE | ID: mdl-34776983

ABSTRACT

Müller cells are the main macroglial cells of the retina exerting a wealth of functions to maintain retinal homoeostasis. Upon pathological changes in the retina, they become gliotic with both protective and detrimental consequences. Accumulating data also provide evidence for a pivotal role of Müller cells in the pathogenesis of diabetic retinopathy (DR). While microglial cells, the resident immune cells of the retina are considered as main players in inflammatory processes associated with DR, the implication of activated Müller cells in chronic retinal inflammation remains to be elucidated. In order to assess the signaling capacity of Müller cells and their role in retinal inflammation, we performed in-depth proteomic analysis of Müller cell proteomes and secretomes after stimulation with INFγ, TNFα, IL-4, IL-6, IL-10, VEGF, TGFß1, TGFß2 and TGFß3. We used both, primary porcine Müller cells and the human Müller cell line MIO-M1 for our hypothesis generating approach. Our results point towards an intense signaling capacity of Müller cells, which reacted in a highly discriminating manner upon treatment with different cytokines. Stimulation of Müller cells resulted in a primarily pro-inflammatory phenotype with secretion of cytokines and components of the complement system. Furthermore, we observed evidence for mitochondrial dysfunction, implying oxidative stress after treatment with the various cytokines. Finally, both MIO-M1 cells and primary porcine Müller cells showed several characteristics of atypical antigen-presenting cells, as they are capable of inducing MHC class I and MHC class II with co-stimulatory molecules. In line with this, they express proteins associated with formation and maturation of phagosomes. Thus, our findings underline the importance of Müller cell signaling in the inflamed retina, indicating an active role in chronic retinal inflammation.

8.
Int J Mol Sci ; 22(17)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34502128

ABSTRACT

Age-related macular degeneration (AMD), one of the leading causes of blindness worldwide, causes personal suffering and high socioeconomic costs. While there has been progress in the treatments for the neovascular form of AMD, no therapy is yet available for the more common dry form, also known as geographic atrophy. We analysed the retinal tissue in a mouse model of retinal degeneration caused by sodium iodate (NaIO3)-induced retinal pigment epithelium (RPE) atrophy to understand the underlying pathology. RNA sequencing (RNA-seq), qRT-PCR, Western blot, immunohistochemistry of the retinas and multiplex ELISA of the mouse serum were applied to find the pathways involved in the degeneration. NaIO3 caused patchy RPE loss and thinning of the photoreceptor layer. This was accompanied by the increased retinal expression of complement components c1s, c3, c4, cfb and cfh. C1s, C3, CFH and CFB were complement proteins, with enhanced deposition at day 3. C4 was upregulated in retinal degeneration at day 10. Consistently, the transcript levels of proinflammatory ccl-2, -3, -5, il-1ß, il-33 and tgf-ß were increased in the retinas of NaIO3 mice, but vegf-a mRNA was reduced. Macrophages, microglia and gliotic Müller cells could be a cellular source for local retinal inflammatory changes in the NaIO3 retina. Systemic complement and cytokines/chemokines remained unaltered in this model of NaIO3-dependent retinal degeneration. In conclusion, systemically administered NaIO3 promotes degenerative and inflammatory processes in the retina, which can mimic the hallmarks of geographic atrophy.


Subject(s)
Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Susceptibility , Iodates/adverse effects , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Complement System Proteins/genetics , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Immunity, Innate , Immunohistochemistry , Mice , Retinal Degeneration/pathology
9.
Int J Mol Sci ; 22(3)2021 Jan 25.
Article in English | MEDLINE | ID: mdl-33503976

ABSTRACT

Müller cells, the major retinal macroglia, are key to maintaining vascular integrity as well as retinal fluid and ion homeostasis. Although platelet derived growth factor (PDGF) receptor expression in Müller glia has been reported earlier, their actual role for Müller cell function and intimate interaction with cells of the retinal neurovascular unit remains unclear. To close this gap of knowledge, Müller cell-specific PDGF receptor alpha (PDGFRα) knockout (KO) mice were generated, characterized, and subjected to a model of choroidal neovascularization (CNV). PDGFRα-deficient Müller cells could not counterbalance hypoosmotic stress as efficiently as their wildtype counterparts. In wildtypes, the PDGFRα ligand PDGF-BB prevented Müller cell swelling induced by the administration of barium ions. This effect could be blocked by the PDGFR family inhibitor AC710. PDGF-BB could not restore the capability of an efficient volume regulation in PDGFRα KO Müller cells. Additionally, PDGFRα KO mice displayed reduced rod and cone-driven light responses. Altogether, these findings suggest that Müller glial PDGFRα is central for retinal functions under physiological conditions. In contrast, Müller cell-specific PDGFRα KO resulted in less vascular leakage and smaller lesion areas in the CNV model. Of note, the effect size was comparable to pharmacological blockade of PDGF signaling alone or in combination with anti-vascular endothelial growth factor (VEGF) therapy-a treatment regimen currently being tested in clinical trials. These data imply that targeting PDGF to treat retinal neovascular diseases may have short-term beneficial effects, but may elicit unwarranted side effects given the putative negative effects on Müller cell homeostatic functions potentially interfering with a long-term positive outcome.


Subject(s)
Ependymoglial Cells/metabolism , Homeostasis , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , Animals , Biomarkers , Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression , Gene Knockout Techniques , Gliosis/etiology , Gliosis/metabolism , Gliosis/pathology , Mice , Mice, Knockout , Neuroglia/metabolism , Organ Specificity/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Retina/metabolism , Retina/pathology
10.
Int J Mol Sci ; 21(22)2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33187113

ABSTRACT

Stargardt macular degeneration is an inherited retinal disease caused by mutations in the ATP-binding cassette subfamily A member 4 (ABCA4) gene. Here, we characterized the complement expression profile in ABCA4-/- retinae and aligned these findings with morphological markers of retinal degeneration. We found an enhanced retinal pigment epithelium (RPE) autofluorescence, cell loss in the inner retina of ABCA4-/- mice and demonstrated age-related differences in complement expression in various retinal cell types irrespective of the genotype. However, 24-week-old ABCA4-/- mice expressed more c3 in the RPE and fewer cfi transcripts in the microglia compared to controls. At the protein level, the decrease of complement inhibitors (complement factor I, CFI) in retinae, as well as an increased C3b/C3 ratio in the RPE/choroid and retinae of ABCA4-/-, mice was confirmed. We showed a corresponding increase of the C3d/C3 ratio in the serum of ABCA4-/- mice, while no changes were observed for CFI. Our findings suggest an overactive complement cascade in the ABCA4-/- retinae that possibly contributes to pathological alterations, including microglial activation and neurodegeneration. Overall, this underpins the importance of well-balanced complement homeostasis to maintain retinal integrity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Complement System Proteins/metabolism , Stargardt Disease/metabolism , Animals , Choroid/metabolism , Complement Activation/physiology , Disease Models, Animal , Female , Macular Degeneration/metabolism , Male , Mice , Mice, Inbred BALB C , Microglia/metabolism , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism
11.
J Neuroinflammation ; 17(1): 1, 2020 Jan 03.
Article in English | MEDLINE | ID: mdl-31900165

ABSTRACT

BACKGROUND: Age-related macular degeneration is characterized by the accumulation of subretinal macrophages and the degeneration of cones, but mainly of rods. We have previously shown that Mononuclear Phagocytes-derived IL-1ß induces rod photoreceptor cell death during experimental subretinal inflammation and in retinal explants exposed to IL-1ß but the mechanism is unknown. METHODS: Retinal explants were culture in the presence of human monocytes or IL-1ß and photoreceptor cell survival was analyzed by TUNEL labeling. Glutamate concentration and transcription levels of gene involved in the homeostasis of glutamate were analyzed in cell fractions of explant cultured or not in the presence of IL-1ß. Glutamate receptor antagonists were evaluated for their ability to reduce photoreceptor cell death in the presence of IL1-ß or monocytes. RESULTS: We here show that IL-1ß does not induce death in isolated photoreceptors, suggesting an indirect effect. We demonstrate that IL-1ß leads to glutamate-induced rod photoreceptor cell death as it increases the extracellular glutamate concentrations in the retina through the inhibition of its conversion to glutamine in Müller cells, increased release from Müller cells, and diminished reuptake. The inhibition of non-NMDA receptors completely and efficiently prevented rod apoptosis in retinal explants cultured in the presence of IL-1ß or, more importantly, in vivo, in a model of subretinal inflammation. CONCLUSIONS: Our study emphasizes the importance of inflammation in the deregulation of glutamate homeostasis and provides a comprehensive mechanism of action for IL-1ß-induced rod degeneration.


Subject(s)
Glutamic Acid/metabolism , Homeostasis/physiology , Interleukin-1beta/toxicity , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Coculture Techniques , Homeostasis/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/drug effects , Monocytes/metabolism , Retinal Rod Photoreceptor Cells/drug effects
12.
Graefes Arch Clin Exp Ophthalmol ; 258(2): 221-230, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31734719

ABSTRACT

Diabetic retinopathy (DR) is a sight-threatening complication associated with the highly prevalent diabetes disorder. Both the microvascular damage and neurodegeneration detected in the retina caused by chronic hyperglycemia have brought special attention to Müller cells, the major macroglia of the retina that are responsible for retinal homeostasis. Given the role of glucocorticoid signaling in anti-inflammatory responses and the almost exclusive expression of glucocorticoid receptors (GRs) in retinal Müller cells, administration of corticosteroid agonists as a potential treatment option has been widely studied. Although these approaches have been moderately efficacious in treating or de-escalating DR pathomechanisms, there are various side effects and gaps of knowledge with regard to introducing exogenous glucocorticoids to the diseased retina. In this paper, we provide a review of the literature concerning the available evidence for the role of Müller cell glucocorticoid signaling in DR and we discuss previously investigated approaches in modulating this system as possible treatment options. Furthermore, we propose a novel alternative to the available choices of treatment by using gene therapy as a tool to regulate the expression of GR in retinal Müller cells. Upregulating GR expression allows for induced glucocorticoid signaling with more enduring effects compared to injection of agonists. Hence, repetitive injections would no longer be required. Lastly, side effects of glucocorticoid therapy such as glucocorticoid resistance of GR following chronic exposure to excess ligands or agonists can be avoided.


Subject(s)
Diabetic Retinopathy/metabolism , Ependymoglial Cells/metabolism , Receptors, Glucocorticoid/metabolism , Retina/metabolism , Animals , Diabetic Retinopathy/pathology , Humans , Retina/pathology , Signal Transduction
13.
Cell Rep ; 29(9): 2835-2848.e4, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31775049

ABSTRACT

Complement dysregulation is a feature of many retinal diseases, yet mechanistic understanding at the cellular level is limited. Given this knowledge gap about which retinal cells express complement, we performed single-cell RNA sequencing on ∼92,000 mouse retinal cells and validated our results in five major purified retinal cell types. We found evidence for a distributed cell-type-specific complement expression across 11 cell types. Notably, Müller cells are the major contributor of complement activators c1s, c3, c4, and cfb. Retinal pigment epithelium (RPE) mainly expresses cfh and the terminal complement components, whereas cfi and cfp transcripts are most abundant in neurons. Aging enhances c1s, cfb, cfp, and cfi expression, while cfh expression decreases. Transient retinal ischemia increases complement expression in microglia, Müller cells, and RPE. In summary, we report a unique complement expression signature for murine retinal cell types suggesting a well-orchestrated regulation of local complement expression in the retinal microenvironment.


Subject(s)
Complement System Proteins/metabolism , Retina/physiopathology , Animals , Humans , Mice
14.
J Neuroinflammation ; 16(1): 43, 2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30777091

ABSTRACT

BACKGROUND: Ligand-driven modulation of the mitochondrial translocator protein 18 kDa (TSPO) was recently described to dampen the neuroinflammatory response of microglia in a retinal light damage model resulting in protective effects on photoreceptors. We characterized the effects of the TSPO ligand XBD173 in the postischemic retina focusing on changes in the response pattern of the major glial cell types of the retina-microglia and Müller cells. METHODS: Retinal ischemia was induced by increasing the intraocular pressure for 60 min followed by reperfusion of the tissue in mice. On retinal cell types enriched via immunomagnetic separation expression analysis of TSPO, its ligand diazepam-binding inhibitor (DBI) and markers of glial activation were performed at transcript and protein level using RNA sequencing, qRT-PCR, lipid chromatography-mass spectrometry, and immunofluorescent labeling. Data on cell morphology and numbers were assessed in retinal slice and flatmount preparations. The retinal functional integrity was determined by electroretinogram recordings. RESULTS: We demonstrate that TSPO is expressed by Müller cells, microglia, vascular cells, retinal pigment epithelium (RPE) of the healthy and postischemic retina, but only at low levels in retinal neurons. While an alleviated neurodegeneration upon XBD173 treatment was found in postischemic retinae as compared to vehicle controls, this neuroprotective effect of XBD173 is mediated putatively by its action on retinal glia. After transient ischemia, TSPO as a marker of activation was upregulated to similar levels in microglia as compared to their counterparts in healthy retinae irrespective of the treatment regimen. However, less microglia were found in XBD173-treated postischemic retinae at 3 days post-surgery (dps) which displayed a more ramified morphology than in retinae of vehicle-treated mice indicating a dampened microglia activation. Müller cells, the major retinal macroglia, show upregulation of the typical gliosis marker GFAP. Importantly, glutamine synthetase was more stably expressed in Müller glia of XBD173-treated postischemic retinae and homeostatic functions such as cellular volume regulation typically diminished in gliotic Müller cells remained functional. CONCLUSIONS: In sum, our data imply that beneficial effects of XBD173 treatment on the postischemic survival of inner retinal neurons were primarily mediated by stabilizing neurosupportive functions of glial cells.


Subject(s)
Ischemia/pathology , Purines/therapeutic use , Retinal Diseases/drug therapy , Retinal Diseases/pathology , Retinal Neurons/drug effects , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Arginase/genetics , Arginase/metabolism , Carrier Proteins/metabolism , Disease Models, Animal , Electroretinography , Gene Expression Regulation/physiology , Glutamate-Ammonia Ligase/metabolism , Ischemia/complications , Ischemia/drug therapy , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuroglia/pathology , RNA, Messenger/metabolism , Receptors, GABA/metabolism , Retina/metabolism , Retina/pathology , Retinal Diseases/complications , Retinal Neurons/classification , Retinal Neurons/pathology , Rhodopsin/metabolism
15.
Elife ; 72018 08 03.
Article in English | MEDLINE | ID: mdl-30074479

ABSTRACT

The P2X7 channel is involved in the pathogenesis of various CNS diseases. An increasing number of studies suggest its presence in neurons where its putative functions remain controversial for more than a decade. To resolve this issue and to provide a model for analysis of P2X7 functions, we generated P2X7 BAC transgenic mice that allow visualization of functional EGFP-tagged P2X7 receptors in vivo. Extensive characterization of these mice revealed dominant P2X7-EGFP protein expression in microglia, Bergmann glia, and oligodendrocytes, but not in neurons. These findings were further validated by microglia- and oligodendrocyte-specific P2X7 deletion and a novel P2X7-specific nanobody. In addition to the first quantitative analysis of P2X7 protein expression in the CNS, we show potential consequences of its overexpression in ischemic retina and post-traumatic cerebral cortex grey matter. This novel mouse model overcomes previous limitations in P2X7 research and will help to determine its physiological roles and contribution to diseases.


Subject(s)
Cerebral Cortex/metabolism , Neurons/metabolism , Purinergic P2X Receptor Antagonists/administration & dosage , Receptors, Purinergic P2X7/genetics , Adenosine Triphosphate/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/genetics , Central Nervous System Diseases/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Disease Models, Animal , Green Fluorescent Proteins/chemistry , Humans , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology
16.
Neurosci Lett ; 670: 69-74, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29391217

ABSTRACT

Leukemia inhibitory factor (LIF) is a cytokine that exerts different effects in the nervous system. It is involved in neuronal injuries and diseases and is assumed to be neuroprotective and to regulate reactive gliosis. In LIF-deficient (LIF-/-) mice, expression of glial fibrillary acidic protein in retinal Müller glial cells as a hallmark of reactive gliosis is suppressed during retinal degenerations. Here, we detected expression of LIF and its receptors in Müller cells of the murine retina. Moreover, electrophysiological alterations of Müller cells 7 days after transient retinal ischemia were studied by the patch-clamp technique. The amplitude of inward currents in Müller cells from the postischemic retina was reduced to 51% in wild type and to 70% in LIF-/- mice. This demonstrates that decrease of inward currents takes place in reactive Müller cells even in the absence of LIF.


Subject(s)
Ependymoglial Cells/physiology , Ischemia/physiopathology , Leukemia Inhibitory Factor/metabolism , Retina/physiopathology , Retinal Vessels/physiopathology , Animals , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Ischemia/metabolism , Ischemia/pathology , Leukemia Inhibitory Factor/genetics , Membrane Potentials/physiology , Mice , Mice, Knockout , Retina/metabolism , Retina/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology
17.
Front Mol Neurosci ; 10: 197, 2017.
Article in English | MEDLINE | ID: mdl-28676742

ABSTRACT

Background: Photoreceptor cell death due to extensive light exposure and induced oxidative-stress are associated with retinal degeneration. A correlated dysregulation of the complement system amplifies the damaging effects, but the local and time-dependent progression of this mechanism is not thoroughly understood. Methods: Light-induced photoreceptor damage (LD) was induced in Balb/c mice with white light illumination either for 24 h with 1000 lux (constant model) or 0.5 h with 5000 lux (acute model). Complement protein and mRNA expression levels were compared at 1 and 3 days post-LD for C1s, complement factor B (CFB), mannose binding lectin A, mannose-binding protein-associated serine protease 1 (MASP-1), C3, C4, C9, and complement factor P in retina and RPE/choroid. Histological analyses visualized apoptosis, microglia/macrophage migration, gliosis and deposition of the complement activation marker C3d. Systemic anaphylatoxin serum concentrations were determined using an ELISA. Results: Apoptosis, gliosis and microglia/macrophage migration into the outer nuclear layer showed similar patterns in both models. Local complement factor expression revealed an early upregulation of complement factor mRNA in the acute and constant light regimen at 1 day post-treatment for c1s, cfb, masp-1, c3, c4 and c9 in the RPE/choroid. However, intraretinal complement mRNA expression for c1s, cfb, c3 and c4 was increased at 1 day in the constant and at 3 days in the acute model. A corresponding regulation on protein level in the retina following both LD models was observed for C3, which was upregulated at 1 day and correlated with increased C3d staining in the ganglion cell layer and at the RPE. In the RPE/choroid C1s-complex protein detection was increased at 3 days after LD irrespectively of the light intensities used. Conclusion: LD in mouse eyes is correlated with local complement activity. The time-dependent local progression of complement regulation on mRNA and protein levels were equivalent in the acute and constant LD model, except for the intraretinal, time-dependent mRNA expression. Knowing the relative time courses of local complement expression and cellular activity can help to elucidate novel therapeutic options in retinal degeneration indicating at which time point of disease complement has to be rebalanced.

18.
Glia ; 65(7): 1059-1071, 2017 07.
Article in English | MEDLINE | ID: mdl-28370368

ABSTRACT

Nervous tissue is characterized by a tight structural association between glial cells and neurons. It is well known that glial cells support neuronal functions, but their role under pathologic conditions is less well understood. Here, we addressed this question in vivo using an experimental model of retinal ischemia and transgenic mice for glia-specific inhibition of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-dependent exocytosis. Transgene expression reduced glutamate, but not ATP release from single Müller cells, impaired glial volume regulation under normal conditions and reduced neuronal dysfunction and death in the inner retina during the early stages of ischemia. Our study reveals that the SNARE-dependent exocytosis in glial cells contributes to neurotoxicity during ischemia in vivo and suggests glial exocytosis as a target for therapeutic approaches.


Subject(s)
Exocytosis/genetics , Ischemia/complications , Nerve Degeneration/etiology , Retina/pathology , Retinal Ganglion Cells/metabolism , SNARE Proteins/metabolism , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Doxycycline/therapeutic use , Ependymoglial Cells/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Intermediate Filaments/metabolism , Ischemia/pathology , Light , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Protein Kinase C-alpha/metabolism , Receptors, Purinergic P2Y1/deficiency , Receptors, Purinergic P2Y1/genetics , SNARE Proteins/genetics , Vascular Endothelial Growth Factor A/metabolism
19.
Front Immunol ; 7: 542, 2016.
Article in English | MEDLINE | ID: mdl-27965669

ABSTRACT

The human complement factor H-related protein-3 (FHR-3) is a soluble regulator of the complement system. Homozygous cfhr3/1 deletion is a genetic risk factor for the autoimmune form of atypical hemolytic-uremic syndrome (aHUS), while also found to be protective in age-related macular degeneration (AMD). The precise function of FHR-3 remains to be fully characterized. We generated four mouse monoclonal antibodies (mAbs) for FHR-3 (RETC) without cross-reactivity to the complement factor H (FH)-family. These antibodies detected FHR-3 from human serum with a mean concentration of 1 µg/mL. FHR-3 levels in patients were significantly increased in sera from systemic lupus erythematosus, rheumatoid arthritis, and polymyalgia rheumatica but remained almost unchanged in samples from AMD or aHUS patients. Moreover, by immunostaining of an aged human donor retina, we discovered a local FHR-3 production by microglia/macrophages. The mAb RETC-2 modulated FHR-3 binding to C3b but not the binding of FHR-3 to heparin. Interestingly, FHR-3 competed with FH for binding C3b and the mAb RETC-2 reduced the interaction of FHR-3 and C3b, resulting in increased FH binding. Our results unveil a previously unknown systemic involvement of FHR-3 in rheumatoid diseases and a putative local role of FHR-3 mediated by microglia/macrophages in the damaged retina. We conclude that the local FHR-3/FH equilibrium in AMD is a potential therapeutic target, which can be modulated by our specific mAb RETC-2.

20.
Neurochem Res ; 41(10): 2598-2606, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27278757

ABSTRACT

Water accumulation in retinal glial (Müller) and neuronal cells resulting in cellular swelling contributes to the development of retinal edema and neurodegeneration. Here, we show that endothelin-1 (ET-1) dose-dependently inhibits the hypoosmotic swelling of Müller cells in freshly isolated retinal slices of control and diabetic rats, with a maximal inhibition at 100 nM. Osmotic Müller cell swelling was also inhibited by ET-2. The effect of ET-1 was mediated by activation of ETA and ETB receptors resulting in transactivation of metabotropic glutamate receptors, purinergic P2Y1, and adenosine A1 receptors. ET-1 (but not ET-2) also inhibited the osmotic swelling of bipolar cells in retinal slices, but failed to inhibit the swelling of freshly isolated bipolar cells. The inhibitory effect of ET-1 on the bipolar cell swelling in retinal slices was abrogated by inhibitors of the FGF receptor kinase (PD173074) and of TGF-ß1 superfamily activin receptor-like kinase receptors (SB431542), respectively. Both Müller and bipolar cells displayed immunoreactivities of ETA and ETB receptor proteins. The data may suggest that neuroprotective effects of ETs in the retina are in part mediated by prevention of the cytotoxic swelling of retinal glial and bipolar cells. ET-1 acts directly on Müller cells, while the inhibitory effect of ET-1 on bipolar cell swelling is indirectly mediated, via stimulation of the release of growth factors like bFGF and TGF-ß1 from Müller cells.


Subject(s)
Endothelins/pharmacology , Ependymoglial Cells/drug effects , Neuroglia/drug effects , Osmosis/drug effects , Retina/drug effects , Signal Transduction/drug effects , Animals , Cell Size/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Ependymoglial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neuroglia/metabolism , Osmotic Pressure/drug effects , Rats, Long-Evans , Rats, Sprague-Dawley , Retina/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...