Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Vis ; 26: 135-149, 2020.
Article in English | MEDLINE | ID: mdl-32180679

ABSTRACT

Purpose: To analyze whether activation of endogenous wingless (Wnt)/ß-catenin signaling in Müller cells is involved in protection of retinal ganglion cells (RGCs) following excitotoxic damage. Methods: Transgenic mice with a tamoxifen-dependent ß-catenin deficiency in Müller cells were injected with N-methyl-D-aspartate (NMDA) into the vitreous cavity of one eye to induce excitotoxic damage of the RGCs, while the contralateral eye received PBS only. Retinal damage was quantified by counting the total number of RGC axons in cross sections of optic nerves and measuring the thickness of the retinal layers on meridional sections. Then, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay was performed to identify apoptotic cells in retinas of both genotypes. Western blot analyses to assess the level of retinal ß-catenin and real-time RT-PCR to quantify the retinal expression of neuroprotective factors were performed. Results: Following NMDA injection of wild-type mice, a statistically significant increase in retinal ß-catenin protein levels was observed compared to PBS-injected controls, an effect that was blocked in mice with a Müller cell-specific ß-catenin deficiency. Furthermore, in mice with a ß-catenin deficiency in Müller cells, NMDA injection led to a statistically significant decrease in RGC axons as well as a substantial increase in TUNEL-positive cells in the RGC layer compared to the NMDA-treated controls. Moreover, in the retinas of the control mice a NMDA-mediated statistically significant induction of leukemia inhibitory factor (Lif) mRNA was detected, an effect that was substantially reduced in mice with a ß-catenin deficiency in Müller cells. Conclusions: Endogenous Wnt/ß-catenin signaling in Müller cells protects RGCs against excitotoxic damage, an effect that is most likely mediated via the induction of neuroprotective factors, such as Lif.


Subject(s)
Ependymoglial Cells/metabolism , Optic Nerve/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism , Tamoxifen/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Apoptosis/drug effects , Axons/drug effects , Axons/metabolism , Ependymoglial Cells/drug effects , In Situ Nick-End Labeling , Leukemia Inhibitory Factor/metabolism , Mice , Mice, Transgenic , N-Methylaspartate/toxicity , Optic Nerve/drug effects , Retina/drug effects , Retina/pathology , Retinal Ganglion Cells/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/deficiency
2.
Invest Ophthalmol Vis Sci ; 59(6): 2240-2251, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29715368

ABSTRACT

Purpose: Norrin is essential for the formation of the retinal vasculature during development and promotes its repair after damage via activation of Wnt/ß-catenin signaling. Since retinal TGF-ß signaling has essentially opposite effects on the retinal vasculature we investigated if and how Norrin inhibits TGF-ß signaling, and vice versa. Methods: Eyes from transgenic mice with an overexpression of Norrin (ßB1-Norrin) and/or active TGF-ß (ßB1-TGF-ß1) in the lens were generated and analyzed by light microscopy, immunohistochemistry, and TUNEL. Further on, protein as well as mRNA levels were investigated by Western blot analyses and real-time RT-PCR, respectively. Results: In ßB1-TGF-ß1 mice, the lack of retinal vascular development and choriocapillaris maintenance was rescued when transgenic Norrin was additionally overexpressed in the eye. In addition, retinal Wnt/ß-catenin signaling and the levels of SMAD7, an inhibitor of the canonical TGF-ß pathway, were substantially suppressed in retinae of ßB1-TGF-ß1 mice. In contrast, Norrin normalized Wnt/ß-catenin signaling and SMAD7 levels in double transgenic mice. Moreover, in retinae of ßB1-TGF-ß1 mice, the amounts of phosphorylated SMAD3, a downstream mediator of TGF-ß signaling, were increased compared to those of ßB1-Norrin/ßB1-TGF-ß1 mice. In vitro, Norrin substantially reduced the TGF-ß-mediated induction of target genes, an effect that was blocked by Dickkopf-1, a specific inhibitor of Wnt/ß-catenin signaling. Conclusions: High amounts of TGF-ß in the eye cause a substantial reduction in the activity of Wnt/ß-catenin signaling. This effect is inhibited in the presence of high amounts of Norrin, which further induce the expression of SMAD7 to inhibit TGF-ß signaling.


Subject(s)
Choroid/metabolism , DNA/genetics , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Retina/metabolism , Transforming Growth Factor beta/genetics , Animals , Blotting, Western , Cell Survival , Cells, Cultured , Choroid/growth & development , Eye Proteins/biosynthesis , Humans , Immunohistochemistry , Mice, Transgenic , Models, Animal , Nerve Tissue Proteins/biosynthesis , Real-Time Polymerase Chain Reaction , Transforming Growth Factor beta/biosynthesis
3.
Sci Rep ; 7(1): 14274, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29079753

ABSTRACT

Norrin is a secreted signaling molecule activating the Wnt/ß-catenin pathway. Since Norrin protects retinal neurons from experimental acute injury, we were interested to learn if Norrin attenuates chronic damage of retinal ganglion cells (RGC) and their axons in a mouse model of glaucoma. Transgenic mice overexpressing Norrin in the retina (Pax6-Norrin) were generated and crossed with DBA/2J mice with hereditary glaucoma and optic nerve axonal degeneration. One-year old DBA/2J/Pax6-Norrin animals had significantly more surviving optic nerve axons than their DBA/2J littermates. The protective effect correlated with an increase in insulin-like growth factor (IGF)-1 mRNA and an enhanced Akt phosphorylation in DBA/2J/Pax6-Norrin mice. Both mouse strains developed an increase in intraocular pressure during the second half of the first year and marked degenerative changes in chamber angle, ciliary body and iris structure. The degenerations were slightly attenuated in the chamber angle of DBA/2J/Pax6-Norrin mice, which showed a ß-catenin increase in the trabecular meshwork. We conclude that high levels of Norrin and the subsequent constitutive activation of Wnt/ß-catenin signaling in RGC protect from glaucomatous axonal damage via IGF-1 causing increased activity of PI3K-Akt signaling. Our results identify components of a protective signaling network preventing degeneration of optic nerve axons in glaucoma.


Subject(s)
Axons/pathology , Eye Proteins/metabolism , Glaucoma/metabolism , Glaucoma/pathology , Nerve Tissue Proteins/metabolism , Optic Nerve/pathology , Animals , Disease Models, Animal , Insulin-Like Growth Factor I/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Signal Transduction
4.
Adv Exp Med Biol ; 801: 347-53, 2014.
Article in English | MEDLINE | ID: mdl-24664717

ABSTRACT

The animal model of N-methyl-D-aspartate (NMDA)-induced excitotoxic damage of retinal ganglion cells (RGC) is widely used to study the molecular mechanisms of RGC death and/or its prevention by neuroprotective agents. NMDA is typically applied by intravitreal injection, while contralateral control eyes are treated by the injection of PBS as vehicle. Herein we report that the procedure of an intravitreal injection alone is sufficient to cause substantial reactive changes in Müller cells and microglia throughout the entire retina. Six week old CD1 mice received a single intravitreal injection of PBS or NMDA. Immunohistochemistry showed the presence of reactive microglia and Müller cells in both NMDA- and PBS-treated eyes during the first 24 h after injection. After 7 days, the reactive changes were only present in NMDA-injected eyes, but no longer in PBS-treated eyes. Investigators using intravitreal injections in the mouse eye should be aware that vehicle-injected control eyes will undergo phenotypic changes in microglia and Müller glia, and are likely to behave differently in their biology when compared with uninjected eyes, at least within the first 24 h after experiment.


Subject(s)
Ependymoglial Cells/drug effects , Gliosis/chemically induced , Microglia/drug effects , N-Methylaspartate/toxicity , Retinal Diseases/chemically induced , Animals , Disease Models, Animal , Ependymoglial Cells/pathology , Excitatory Amino Acid Agonists/toxicity , Gliosis/pathology , Intravitreal Injections , Mice , Mice, Inbred Strains , Microglia/pathology , Retina/drug effects , Retina/pathology , Retinal Diseases/pathology
5.
Cell Tissue Res ; 353(2): 339-45, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23779255

ABSTRACT

Cells of Müller glia and microglia react to neuronal injury in glaucoma. The change to a reactive phenotype initiates signaling cascades that may serve a neuroprotective role, but may also proceed to promote damaging effects on retinal neurons. Both effects appear to occur most likely in parallel in glaucoma, but the underlying mechanisms and signaling pathways that specifically promote protective versus destructive roles of reactive glial cells are mostly unclear. More research is needed to understand the homeostatic signaling network in which retinal glia cells are embedded to maintain or restore neuronal function after injury.


Subject(s)
Ependymoglial Cells/pathology , Glaucoma/pathology , Microglia/pathology , Animals , Health , Humans
6.
Methods Mol Biol ; 935: 99-109, 2013.
Article in English | MEDLINE | ID: mdl-23150363

ABSTRACT

The animal model of N-methyl-D-aspartate (NMDA)-induced excitotoxic damage of retinal ganglion cells (RGC) is widely used to study the molecular mechanisms of RGC apoptosis and/or its prevention by neuroprotective agents. This chapter provides protocols for applying NMDA-induced excitotoxic damage to RGC of mouse eyes and for subsequent measuring of the extent of the resulting damage.


Subject(s)
Disease Models, Animal , Excitatory Amino Acid Agonists , Eye Diseases/chemically induced , Mice , N-Methylaspartate , Retinal Ganglion Cells/pathology , Animals , Apoptosis/drug effects , Eye/cytology , Eye/pathology , Eye Diseases/pathology , Neuroprotective Agents/pharmacology
7.
Histochem Cell Biol ; 138(5): 709-24, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22782339

ABSTRACT

Plasmalemmal vesicle-associated protein (PLVAP, PV-1) is specifically expressed in endothelial cells in which it localizes to diaphragms of fenestrae, caveolae, and transendothelial channels. To learn about its function, we generated mutant mice that lack PLVAP. In a C57BL/6N genetic background, homozygous Plvap-deficient embryos die before birth and suffer from subcutaneous edema, hemorrhages, and defects in the vascular wall of subcutaneous capillaries. In addition, hearts of Plvap(-/-) embryos show ventricular septal defects and thinner ventricular walls. In wild-type embryos, PLVAP and caveolae with a stomatal diaphragm are present in endothelial cells of subcutaneous capillaries and endocardium, while a diaphragm is missing in caveolae of Plvap(-/-) littermates. Plvap(-/-) mice in a mixed C57BL/6N/FVB-N genetic background are born and survive at the most for 4 weeks. Capillaries of exocrine and endocrine pancreas and of kidney peritubular interstitium were investigated in more detail as examples of fenestrated capillaries. In these vascular beds, Plvap(-/-) mice show a complete absence of diaphragms in fenestrae, caveolae, and transendothelial channels, findings which are associated with a substantial decrease in the number of endothelial fenestrae. The changes in the capillary phenotype correlate with a considerable retardation of postnatal growth and anemia. Plvap(-/-) mice provide an animal model to clarify the specific functional role of endothelial fenestrae and their contribution to passage of water and solutes in different organs.


Subject(s)
Carrier Proteins/genetics , Endothelium, Vascular/abnormalities , Membrane Proteins/genetics , Animals , Capillaries/abnormalities , Caveolae , Endocardium/abnormalities , Female , Homozygote , Kidney/abnormalities , Kidney/blood supply , Male , Mice , Mice, Inbred C57BL , Models, Animal , Mutation , Pancreas/abnormalities , Pancreas/blood supply
8.
J Neurosci ; 30(17): 5998-6010, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20427659

ABSTRACT

Norrin is a secreted protein that binds to frizzled 4 and controls development of capillaries in retina and inner ear. We provide evidence that Norrin has distinct neuroprotective properties that are independent from its effects on vascular development. The function of Norrin was investigated in a mouse model of excitotoxic retinal ganglion cell (RGC) damage after intravitreal injection of NMDA, and in cultured Müller glia or immortalized RGC-5 cells. Intravitreal injection of Norrin significantly increased the number of surviving RGC axons in the optic nerve and decreased apoptotic death of retinal neurons following NMDA-mediated damage. This effect could be blocked by adding dickkopf (DKK)-1, an inhibitor of the Wnt/beta-catenin signaling pathway. Treatment of eyes with combined Norrin/NMDA activated Wnt/beta-catenin signaling and increased the retinal expression of leukemia inhibitory factor and endothelin-2, as well as that of neurotrophic growth factors such as fibroblast growth factor-2, brain-derived neurotrophic factor, lens epithelium-derived growth factor, and ciliary neurotrophic factor. A similar activation of Wnt/beta-catenin signaling and an increased expression of neurotrophic factors was observed in cultured Müller cells after treatment with Norrin, effects that again could be blocked by adding DKK-1. In addition, conditioned cell culture medium of Norrin-treated Müller cells increased survival of differentiated RGC-5 cells. We conclude that Norrin has pronounced neuroprotective properties on retinal neurons with the distinct potential to decrease the damaging effects of NMDA-induced RGC loss. The effects of Norrin involve activation of Wnt/beta-catenin signaling and subsequent induction of neurotrophic growth factors in Müller cells.


Subject(s)
Eye Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroglia/physiology , Retinal Ganglion Cells/physiology , Animals , Cell Line , Cell Survival/physiology , Cells, Cultured , Culture Media, Conditioned , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , N-Methylaspartate/metabolism , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism
9.
J Neurosci ; 30(1): 183-93, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053900

ABSTRACT

Norrin is a secreted protein that is involved in retinal angiogenesis and activates the Wnt-signaling pathway. We studied the role of Norrin in microvascular endothelial cells in vitro, and in a mouse model of retinopathy characterized by oxygen-induced vascular loss followed by hypoxia-induced pathological neovascularization. Recombinant Norrin significantly increased proliferation, viability, migration, and tube formation in vitro. Two independent transgenic mouse strains with ectopic overexpression of Norrin from the lens (betaB1-Crystallin-Norrin), or the retinal pigment epithelium (Rpe65-Norrin) were generated and exposed to high oxygen. Following oxygen treatment, vascular loss was significantly smaller in retinae of transgenic mice from both strains as compared to wild-type littermates. In addition, the anatomical correct regrowth of vessels was significantly increased, while pathological neovascularization was suppressed. In vitro and in vivo effects of Norrin could be blocked by adding DKK (Dickkopf)-1, an inhibitor of Wnt/beta-catenin signaling. Treatment of microvascular endothelial cells with Norrin caused a substantial increase in the expression of angiopoietin-2 (Ang-2). When inhibitory antibodies against Ang-2 were added to Norrin, the proliferative effects of Norrin were significantly suppressed. We conclude that Norrin is a potent factor to induce angiogenesis in microvascular endothelial cells, which has the distinct potential to suppress the damaging effects of oxygen-induced retinopathy in vivo. The effects of Norrin appear to be mediated, at least partially, via the induction of Ang-2.


Subject(s)
Eye Proteins/physiology , Nerve Tissue Proteins/physiology , Oxygen/toxicity , Retinal Diseases/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/physiopathology , Retinal Vessels/growth & development , Retinal Vessels/metabolism , Animals , Cell Line , Cell Survival/physiology , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Humans , Mice , Mice, Transgenic , Microcirculation/physiology , Retinal Diseases/physiopathology
10.
Mol Pharmacol ; 71(1): 366-76, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17065237

ABSTRACT

According to previous reports, flavonoids and nutraceuticals correct defective electrolyte transport in cystic fibrosis (CF) airways. Traditional medicinal plants from China and Thailand contain phytoflavonoids and other bioactive compounds. We examined herbal extracts of the common Thai medicinal euphorbiaceous plant Phyllanthus acidus for their potential effects on epithelial transport. Functional assays by Ussing chamber, patch-clamping, double-electrode voltage-clamp and Ca2+ imaging demonstrate activation of Cl- secretion and inhibition of Na+ absorption by P. acidus. No cytotoxic effects of P. acidus could be detected. Mucosal application of P. acidus to native mouse trachea suggested transient and steady-state activation of Cl- secretion by increasing both intracellular Ca2+ and cAMP. These effects were mimicked by a mix of the isolated components adenosine, kaempferol, and hypogallic acid. Additional experiments in human airway cells and CF transmembrane conductance regulator (CFTR)-expressing BHK cells and Xenopus laevis oocytes confirm the results obtained in native tissues. Cl- secretion was also induced in tracheas of CF mice homozygous for Phe508del-CFTR and in Phe508del-CFTR homozygous human airway epithelial cells. Taken together, P. acidus corrects defective electrolyte transport in CF airways by parallel mechanisms including 1) increasing the intracellular levels of second messengers cAMP and Ca2+, thereby activating Ca2+-dependent Cl- channels and residual CFTR-Cl- conductance; 2) stimulating basolateral K+ channels; 3) redistributing cellular localization of CFTR; 4) directly activating CFTR; and 5) inhibiting ENaC through activation of CFTR. These combinatorial effects on epithelial transport may provide a novel complementary nutraceutical treatment for the CF lung disease.


Subject(s)
Chlorides/metabolism , Phyllanthus , Plant Extracts/pharmacology , Plants, Medicinal , Calcium/metabolism , Cell Culture Techniques , Cell Survival , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Sodium Channels/genetics , Patch-Clamp Techniques , Plant Leaves
SELECTION OF CITATIONS
SEARCH DETAIL
...