Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Neurosci ; 40(3): 2502-12, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24754835

ABSTRACT

Toll-like receptor 4 (Tlr4) plays an important role in ischemia-reperfusion (IR)-induced retinal inflammation and damage. However, the role of two Tlr4-dependent signaling cascades, myeloid differentiation primary response 88 (Myd88) and TIR-domain-containing adapter inducing interferon-ß (Trif), in retinal IR injury is poorly understood. In this study, we investigated the contribution of the Myd88-dependent and Trif-dependent signaling cascades in retinal damage and inflammation triggered by IR, by using Myd88 knockout (Myd88KO) and Trif knockout (TrifKO) mice. Retinal IR injury was induced by unilateral elevation of intraocular pressure for 45 min by direct corneal cannulation. To study IR-induced retinal ganglion cell (RGC) death in vitro, we used an oxygen and glucose deprivation (OGD) model. Our data suggested that Myd88 was present in many retinal layers of sham-operated and ischemic mice, whereas Trif was mainly present in the ganglion cell layer (GCL). The level of Myd88 was increased in the retina after IR. We found that retinas of TrifKO mice had a significantly reduced neurotoxic pro-inflammatory response and significantly increased survival of the GCL neurons after IR. Although Myd88KO mice had relatively low levels of inflammation in ischemic retinas, their levels of IR-induced retinal damage were notably higher than those of TrifKO mice. We also found that Trif-deficient RGCs were more resistant to death induced by OGD than were RGCs isolated from Myd88KO mice. These data suggested that, as compared with the Myd88-dependent signaling cascade, Trif signaling contributes significantly to retinal damage after IR.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Myeloid Differentiation Factor 88/metabolism , Retinal Diseases/metabolism , Retinitis/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Reperfusion Injury/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism
2.
J Neuroinflammation ; 11: 81, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24755298

ABSTRACT

BACKGROUND: Sterile inflammation occurs in the absence of live pathogens and is an unavoidable consequence of ischemia-reperfusion (IR) injury in the central nervous system (CNS). It is known that toll-like receptor 4 (Tlr4) contributes to damage and sterile inflammation in the CNS mediated by IR. However, the mechanism of Tlr4 activation under sterile conditions in ischemic tissue is poorly understood. We performed this study to clarify the mechanism. To this end, we focused on the extracellular heat shock protein 70 (Hsp70), the prototypic Tlr4 ligand. METHODS: Tlr4-, Myd88- and Trif-knockout animals, as well as C57BL/6 mice, were used for the wild type control. For the in vivo study, we used a mouse model of retinal IR injury. To test the role of protein kinase C (PKC) in IR injury, IR retinas were treated with the PKC inhibitors (polymyxin B and Gö6976) and retinal damage was evaluated by directly counting neurons in the ganglion cell layer of flat-mounted retinas seven days after IR. Primary retinal neurons (retinal ganglion cells) and glial cells were used for in vitro experiments. Quantitative RT-PCR, ELISA and western blot analysis were used to study the production of pro-inflammatory factors in IR retinas and in primary cell cultures. RESULTS: We found significant accumulation of extracellular Hsp70 in a model of retinal IR injury. We noted that PKC was involved in Tlr4 signaling, and found that PKC inhibitors promoted neuroprotection by reducing pro-inflammatory activity in ischemic tissue. To put all of the pieces in the signaling cascade together, we performed an in vitro study. We found that PKC was critical to mediate the Hsp70-dependent pro-inflammatory response. At the same time, the contamination of Hsp70 preparations with low-dose endotoxin was not critical to mediate the production of pro-inflammatory factors. We found that extracellular Hsp70 can promote neuronal death at least, by mediating production of cytotoxic levels of tumor necrosis factor alpha, predominantly due to the Tlr4/Myd88 signaling cascade. CONCLUSIONS: Our findings suggest that PKC acts as a switch to amplify the pro-inflammatory activity of Hsp70/Tlr4 signaling, which is sufficient to mediate neuronal death.


Subject(s)
Endotoxins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Inflammation/etiology , Protein Kinase C/metabolism , Reperfusion , Retina/metabolism , Retina/pathology , Animals , Cell Count , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Inflammation/metabolism , Ischemia/complications , Ischemia/drug therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Retina/cytology , Retina/drug effects
3.
PLoS One ; 9(1): e87574, 2014.
Article in English | MEDLINE | ID: mdl-24498140

ABSTRACT

Diabetic retinopathy is one of the main microvascular complications of diabetes and remains one of the leading causes of blindness worldwide. Recent studies have revealed an important role of inflammatory and proangiogenic high mobility group 1 (HMGB-1) cytokine in diabetic retinopathy. To elucidate cellular mechanisms of HMGB-1 activity in the retina, we performed this study. The histological features of diabetic retinopathy include loss of blood-vessel pericytes and endothelial cells, as well as abnormal new blood vessel growth. To establish the role of HMGB-1 in vulnerability of endothelial cells and pericytes, cultures of these cells, or co-cultures with glial cells, were treated with HMGB-1 and assessed for survival after 24 hours. The expression levels of the cytokines, chemokines, and cell adhesion molecules in glial and endothelial cells were tested by quantitative RT-PCR to evaluate changes in these cells after HMGB-1 treatment. Animal models of neovascularization were also used to study the role of HMGB-1 in the retina. We report that pericyte death is mediated by HMGB-1-induced cytotoxic activity of glial cells, while HMGB-1 can directly mediate death of endothelial cells. We also found that HMGB-1 affects endothelial cell activity. However, we did not observe a difference in the levels of neovascularization between HMGB-1-treated eyes compared to the control eyes, nor in the levels of proangiogenic cytokine VEGF-A expression between glial cells treated with HMGB-1 and control cells. Our data also indicate that HMGB-1 is not involved in retinal neovascularization in the oxygen-induced retinopathy model. Thus, our data suggest that retinal pericyte and endothelial injury and death in diabetic retinopathy may be due to HMGB-1-induced cytotoxic activity of glial cells as well as the direct effect of HMGB-1 on endothelial cells. At the same time, our findings indicate that HMGB-1 plays an insignificant role in retinal and choroidal neovascularization.


Subject(s)
Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , HMGB1 Protein/metabolism , Retinal Neovascularization/metabolism , Animals , Cells, Cultured , Coculture Techniques , Diabetic Retinopathy/pathology , Endothelial Cells/pathology , Humans , Mice , Neuroglia/metabolism , Neuroglia/pathology , Pericytes/metabolism , Pericytes/pathology , Retinal Neovascularization/pathology
4.
Mol Vis ; 19: 2596-605, 2013.
Article in English | MEDLINE | ID: mdl-24379647

ABSTRACT

PURPOSE: The aim of this study was to develop and characterize a new contact lens-associated fungal keratitis rat model and to assess the ability of non-invasive spectral-domain optical coherence tomography (SD-OCT) to detect pathological changes in vivo in fungal keratitis. METHODS: We used SD-OCT to image and measure the cornea of Sprague Dawley rats. Fusarium infection was initiated in the rat eye by fitting Fusarium solani-soaked contact lenses on the experimental eye, while the control animals received contact lenses soaked in sterile saline. The fungal infection was monitored with periodic slit-lamp examination and in vivo SD-OCT imaging of the rat eye, and confirmed by histology, counting of viable fungi in the infected rat cornea, and PCR with specific primers for Fusarium sp. RESULTS: We imaged and measured the rat cornea with SD-OCT. Custom-made contact lenses were developed based on the OCT measurements. Incubation of contact lenses in a F. solani suspension resulted in biofilm formation. We induced contact lens-associated Fusarium keratitis by fitting the rat eyes for 4 h with the Fusarium-contaminated contact lenses. The SD-OCT images of the cornea correlated well with the slit-lamp and histopathological results and clearly defined clinical signs of infection, namely, increased corneal thickening, loss of epithelial continuity, hyper-reflective areas representing infiltrates, and endothelial plaques characteristic of fungal infection. Moreover, in three cases, SD-OCT detected the infection without any clear findings on slit-lamp examination. Infection was confirmed with histological fungal staining, PCR, and microbiological culture positivity. CONCLUSIONS: We developed a highly reproducible rat contact lens model and successfully induced contact lens-associated Fusarium keratitis in this model. The clinical presentation of contact lens-associated Fusarium keratitis in the rat model is similar to the human condition. SD-OCT is a valuable tool that non-invasively revealed characteristic signs of the fungal infection and could provide sensitive, objective monitoring in fungal keratitis.


Subject(s)
Contact Lenses/microbiology , Eye Infections, Fungal/pathology , Fusariosis/pathology , Keratitis/pathology , Animals , Contact Lens Solutions , Disease Models, Animal , Eye Infections, Fungal/diagnosis , Eye Infections, Fungal/microbiology , Female , Fusariosis/diagnosis , Fusariosis/microbiology , Fusarium/pathogenicity , Fusarium/physiology , Humans , Keratitis/diagnosis , Keratitis/microbiology , Rats , Rats, Sprague-Dawley , Tomography, Optical Coherence
5.
PLoS One ; 7(10): e48332, 2012.
Article in English | MEDLINE | ID: mdl-23118988

ABSTRACT

Extracellular matrix (ECM) integrity in the central nervous system (CNS) is essential for neuronal homeostasis. Signals from the ECM are transmitted to neurons through integrins, a family of cell surface receptors that mediate cell attachment to ECM. We have previously established a causal link between the activation of the matrix metalloproteinase-9 (MMP-9), degradation of laminin in the ECM of retinal ganglion cells (RGCs), and RGC death in a mouse model of retinal ischemia-reperfusion injury (RIRI). Here we investigated the role of laminin-integrin signaling in RGC survival in vitro, and after ischemia in vivo. In purified primary rat RGCs, stimulation of the ß1 integrin receptor with laminin, or agonist antibodies enhanced RGC survival in correlation with activation of ß1 integrin's major downstream regulator, focal adhesion kinase (FAK). Furthermore, ß1 integrin binding and FAK activation were required for RGCs' survival response to laminin. Finally, in vivo after RIRI, we observed an up-regulation of MMP-9, proteolytic degradation of laminin, decreased RGC expression of ß1 integrin, FAK and Akt dephosphorylation, and reduced expression of the pro-survival molecule bcl-xL in the period preceding RGC apoptosis. RGC death was prevented, in the context of laminin degradation, by maintaining ß1 integrin activation with agonist antibodies. Thus, disruption of homeostatic RGC-laminin interaction and signaling leads to cell death after retinal ischemia, and maintaining integrin activation may be a therapeutic approach to neuroprotection.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin beta1/metabolism , Retinal Ganglion Cells/cytology , Signal Transduction , Animals , Apoptosis , Cell Adhesion , Cell Survival , Female , Gene Expression Regulation, Enzymologic , Laminin/metabolism , Matrix Metalloproteinase 9/metabolism , Neurites/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Retinal Ganglion Cells/enzymology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
6.
Invest Ophthalmol Vis Sci ; 53(6): 2823-30, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22467573

ABSTRACT

PURPOSE: To study the role of neuronal nicotinamide adenine dinucleotide phosphate [NAD(P)H] oxidase-dependent reactive oxygen species (ROS) production in retinal ganglion cell (RGC) death after ischemia. METHODS: Ischemic injury was induced by unilateral elevation of intraocular pressure via direct corneal cannulation. For in vitro experiments, RGCs isolated by immunopanning from retinas were exposed to oxygen and glucose deprivation (OGD). The expression levels of NAD(P)H oxidase subunits were evaluated by quantitative PCR, immunocytochemistry, and immunohistochemistry. The level of ROS generated was assayed by dihydroethidium. The NAD(P)H oxidase inhibitors were then tested to determine if inhibition of NAD(P)H oxidase altered the production of ROS within the RGCs and promoted cell survival. RESULTS: It was reported that RGCs express catalytic Nox1, Nox2, Nox4, Duox1, as well as regulatory Ncf1/p47phox, Ncf2/p67phox, Cyba/p22phox, Noxo1, and Noxa1 subunits of NAD(P)H oxidases under normal conditions and after ischemia. However, whereas RGCs express only low levels of catalytic Nox2, Nox4, and Duox1, and regulatory Ncf1/p47, Ncf2/p67 subunits, they exhibit significantly higher levels of catalytic subunit Nox1 and the subunits required for optimal activity of Nox1. It was observed that the nonselective NAD(P)H oxidase inhibitors VAS-2870, AEBSF, and the Nox1 NAD(P)H oxidase-specific inhibitor ML-090 decreased the ROS burst stimulated by OGD, which was associated with a decreased level of RGC death. CONCLUSIONS: The findings suggest that NAD(P)H oxidase activity in RGCs renders them vulnerable to ischemic death. Importantly, high levels of Nox1 NAD(P)H oxidase subunits in RGCs suggest that this enzyme could be a major source of ROS in RGCs produced by NAD(P)H oxidases.


Subject(s)
Cell Death/physiology , Ischemia/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Retinal Ganglion Cells/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Immunohistochemistry , Ischemia/enzymology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , RNA, Messenger/metabolism , Retinal Ganglion Cells/enzymology
7.
Invest Ophthalmol Vis Sci ; 52(10): 7187-94, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21828158

ABSTRACT

PURPOSE: High-mobility group protein B1 (Hmgb1) is released from necrotic cells and induces an inflammatory response. Although Hmgb1 has been implicated in ischemia/reperfusion (IR) injury of the brain, its role in IR injury of the retina remains unclear. Here, the authors provide evidence that Hmgb1 contributes to retinal damage after IR. METHODS: Retinal IR injury was induced by unilateral elevation of intraocular pressure and the level of Hmgb1 in vitreous humor was analyzed 24 hours after reperfusion. To test the functional significance of Hmgb1 release, ischemic or normal retinas were treated with the neutralizing anti-Hmgb1 antibody or recombinant Hmgb1 protein respectively. To elucidate in which cell type Hmgb1 exerts its effect, primary retinal ganglion cell (RGC) cultures and glia RGC cocultures were treated with Hmgb1. To clarify the downstream signaling pathways involved in Hmgb1-induced effects in the ischemic retina, receptor for advanced glycation end products (Rage)-deficient mice (RageKO) were used. RESULTS: Hmgb1 is accumulated in the vitreous humor 24 hours after IR. Inhibition of Hmgb1 activity with neutralizing antibody significantly decreased retinal damage after IR, whereas treatment of retinas or retinal cells with Hmgb1 induced a loss of RGCs. The analysis of RageKO versus wild-type mice showed significantly reduced expression of proinflammatory genes 24 hours after reperfusion and significantly increased survival of ganglion cell layer neurons 7 days after IR injury. CONCLUSIONS: These results suggest that an increased level of Hmgb1 and signaling via the Rage contribute to neurotoxicity after retinal IR injury.


Subject(s)
HMGB1 Protein/physiology , Reperfusion Injury/metabolism , Retinal Diseases/metabolism , Retinal Ganglion Cells/metabolism , Animals , Animals, Newborn , Antibodies, Neutralizing/pharmacology , Blotting, Western , Cell Survival , Cells, Cultured , Coculture Techniques , Fluorescent Antibody Technique, Indirect , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Neuroglia/pathology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Recombinant Proteins/pharmacology , Reperfusion Injury/pathology , Retinal Diseases/pathology , Retinal Ganglion Cells/pathology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...