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










Publication year range
1.
J Clin Med ; 10(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34640509

ABSTRACT

High myopia is a global ocular disease and one of the most common causes of blindness. Fundus images can be obtained in a noninvasive manner and can be used to monitor and follow up on many fundus diseases, such as high myopia. In this paper, we proposed a computer-aided diagnosis algorithm using deep convolutional neural networks (DCNNs) to grade the risk of high myopia. The input images were automatically classified into three categories: normal fundus images were labeled class 0, low-risk high-myopia images were labeled class 1, and high-risk high-myopia images were labeled class 2. We conducted model training on 758 clinical fundus images collected locally, and the average accuracy reached 98.15% according to the results of fivefold cross-validation. An additional 100 fundus images were used to evaluate the performance of DCNNs, with ophthalmologists performing external validation. The experimental results showed that DCNNs outperformed human experts with an area under the curve (AUC) of 0.9968 for the recognition of low-risk high myopia and 0.9964 for the recognition of high-risk high myopia. In this study, we were able to accurately and automatically perform high myopia classification solely using fundus images. This has great practical significance in terms of improving early diagnosis, prevention, and treatment in clinical practice.

2.
Oncotarget ; 7(37): 60123-60132, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27517753

ABSTRACT

Excessive Ultra-violet (UV) radiation causes oxidative damages and apoptosis in retinal pigment epithelium (RPE) cells. Here we tested the potential activity of SC79, a novel small molecule activator of Akt, against the process. We showed that SC79 activated Akt in primary and established (ARPE-19 line) RPE cells. It protected RPE cells from UV damages possibly via inhibiting cell apoptosis. Akt inhibition, via an Akt specific inhibitor (MK-2206) or Akt1 shRNA silence, almost abolished SC79-induced RPE cytoprotection. Further studies showed that SC79 activated Akt-dependent NF-E2-related factor 2 (Nrf2) signaling and inhibited UV-induced oxidative stress in RPE cells. Reversely, Nrf2 shRNA knockdown or S40T mutation attenuated SC79-induced anti-UV activity. For the in vivo studies, we showed that intravitreal injection of SC79 significantly protected mouse retina from light damages. Based on these results, we suggest that SC79 protects RPE cells from UV damages possibly via activating Akt-Nrf2 signaling axis.


Subject(s)
Acetates/pharmacology , Benzopyrans/pharmacology , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinal Pigment Epithelium/drug effects , Signal Transduction/drug effects , Acetates/chemistry , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Benzopyrans/chemistry , Cell Line , Cells, Cultured , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Male , Mice, Inbred BALB C , Molecular Structure , Mutation , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/radiation effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Ultraviolet Rays
3.
Cell Physiol Biochem ; 35(5): 1663-76, 2015.
Article in English | MEDLINE | ID: mdl-25833234

ABSTRACT

Autophagy is an important intracellular degradative process that delivers cytoplasmic proteins to lysosome for degradation. Dysfunction of autophagy is implicated in several human diseases, such as neurodegenerative diseases, infectious diseases, and cancers. Autophagy-related proteins are constitutively expressed in the eye. Increasing studies have revealed that abnormal autophagy is an important pathological feature of several ocular diseases. Pharmacological manipulation of autophagy may provide an alternative therapeutic target for some ocular diseases. In this manuscript, we reviewed the relevant progress about the role of autophagy in the pathogenesis of ocular diseases.


Subject(s)
Autophagy , Eye Diseases/pathology , Cataract/metabolism , Cataract/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Eye Diseases/metabolism , Glaucoma/metabolism , Glaucoma/pathology , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Retinal Pigment Epithelium/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism
4.
Cell Physiol Biochem ; 33(1): 107-16, 2014.
Article in English | MEDLINE | ID: mdl-24481000

ABSTRACT

BACKGROUND: Autophagy is a self-degradative process that is important for balancing sources of energy at critical times in development and in response to nutrient stress. Retinal pigment epithelium (RPE) works as the outer blood retina barrier and is vulnerable to energy stress-induced injury. However, the effect of high glucose treatment on autophagy is still unclear in RPE. METHODS: Transmission electron microscopy was used to detect the generation of autophagosome. Small interfering RNA (siRNA) and MTT was used to determine the effect of autophagy on cell viability. Western blots and immunohistochemistry were used to detect the expression pattern of autophagic markers, including LC3 and p62. RESULTS: High glucose treatment results in a significant increase in the generation of autophagosome and altered expression of LC3 and p62. High glucose-induced autophagy is independent of mTOR signaling, but is mainly regulated via ROS-mediated ER stress signaling. CONCLUSION: In the scenario of high glucose-induced oxidative stress, autophagy may be required for the removal of damaged proteins, and provide a default mechanism to prevent high glucose-induced injury in RPE.


Subject(s)
Autophagy/drug effects , Glucose/pharmacology , Retinal Pigment Epithelium/cytology , Biomarkers/metabolism , Endoplasmic Reticulum Stress/drug effects , Humans , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
5.
Neurol Sci ; 35(6): 847-53, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24368741

ABSTRACT

Glutamate is a major excitatory neurotransmitter in the retina. Glutamate neurotoxicity has been implicated in the pathogenesis of several ocular diseases. Aquaporin 4 (AQP4) is a water-selective membrane transport protein, and its knockout could alter retinal neuron excitability. However, the effect of AQP4 knockout on glutamate metabolism is still unclear in the retina. Here, we reported that the retinas in AQP4 knockout mice showed higher glutamate levels than that in wild-type mice upon light damage. AQP4 knockout could result in accelerated apoptosis of retinal cells, increased reactive gliosis, and attenuated survival of RGCs in response to light damage. Moreover, AQP4 knockout could affect the expression pattern of glutamate metabolism-related proteins such as GLAST and GS. Taken together, this study revealed a novel role of AQP4 in regulating glutamate metabolism. Pharmacological manipulation of AQP4 function may represent as a potent therapeutic target in the treatment of neurological ocular disorders.


Subject(s)
Aquaporin 4/genetics , Glutamic Acid/metabolism , Retina/metabolism , Animals , Female , Light , Mice , Mice, Knockout
6.
Int J Mol Sci ; 14(5): 10355-68, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23685869

ABSTRACT

Ultraviolet (UV) radiation and reactive oxygen species (ROS) impair the physiological functions of retinal pigment epithelium (RPE) cells by inducing cell apoptosis, which is the main cause of age-related macular degeneration (AMD). The mechanism by which UV/ROS induces RPE cell death is not fully addressed. Here, we observed the activation of a ceramide-endoplasmic reticulum (ER) stress-AMP activated protein kinase (AMPK) signaling axis in UV and hydrogen peroxide (H2O2)-treated RPE cells. UV and H2O2 induced an early ceramide production, profound ER stress and AMPK activation. Pharmacological inhibitors against ER stress (salubrinal), ceramide production (fumonisin B1) and AMPK activation (compound C) suppressed UV- and H2O2-induced RPE cell apoptosis. Conversely, cell permeable short-chain C6 ceramide and AMPK activator AICAR (5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide) mimicked UV and H2O2's effects and promoted RPE cell apoptosis. Together, these results suggest that UV/H2O2 activates the ceramide-ER stress-AMPK signaling axis to promote RPE cell apoptosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/physiology , Ceramides/metabolism , Endoplasmic Reticulum Stress/physiology , Hydrogen Peroxide/pharmacology , Ultraviolet Rays , Apoptosis/drug effects , Apoptosis/radiation effects , Blotting, Western , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cell Survival/radiation effects , Ceramides/pharmacology , Cinnamates/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/radiation effects , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Fumonisins/pharmacology , Humans , Oxidants/pharmacology , Retinal Pigment Epithelium/cytology , Signal Transduction/drug effects , Signal Transduction/physiology , Signal Transduction/radiation effects , Thiourea/analogs & derivatives , Thiourea/pharmacology
7.
Exp Ther Med ; 5(2): 621-625, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23403739

ABSTRACT

Nerve distal axon injury-induced Wallerian degeneration is significantly delayed in Wallerian degeneration slow (Wld(s)) mutant mice, although the associated mechanisms are not completely clear and the role of Wld(s) in retinal ganglion cell (RGC) body damage is not fully understood. In the present study, a Wallerian degeneration model was established in wild-type (WT) and Wld(s) mutant mice by creating mechanical injury in the optic nerves. Wallerian degeneration and RGC body collapse were observed to be significantly delayed in the Wld(s) mice. Electroretinograms (ERG) and visual evoked potentials (VEPs) in Wld(s) mice were also significantly improved at the earlier stages (one week) following injury. The retina immunohistochemistry results showed that Wld(s) mice had more ordered cells and improved inner granular cell layer arrangement compared with the WT mice. Optic nerve Luxol Fast Blue (LFB) staining showed greater axon demyelination in WT mice than in Wld(s) mice. A large number of apoptotic cells were also observed in the WT mice. The present results suggest that the Wld(s) gene may also protect the RGC body following nerve injury.

8.
Biochem Biophys Res Commun ; 425(1): 33-8, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22820188

ABSTRACT

Tumor necrosis factor-alpha (TNF-α) promotes in vitro retinal pigment epithelial (RPE) cell migration to initiate proliferative vitreoretinopathy (PVR). Here we report that TNF-α promotes human RPE cell migration by inducing matrix metallopeptidase 9 (MMP-9) expression. Inhibition of MMP-9 by its inhibitor or its neutralizing antibody inhibited TNF-α-induced in vitro RPE cell migration. Reversely, exogenously-added active MMP-9 promoted RPE cell migration. Suppression Akt/mTOR complex 1(mTORC1) activation by LY 294002 and rapamycin inhibited TNF-α-mediated MMP-9 expression. To introduce a constitutively active Akt (CA-Akt) in cultured RPE cells increased MMP-9 expression, and to block mTORC1 activation by rapamycin inhibited its effect. RNA interference (RNAi)-mediated silencing of SIN1, a key component of mTOR complex 2 (mTORC2), had no effect on MMP-9 expression or secretion. In conclusion, this study suggest that TNF-α promotes RPE cell migration by inducing MMP-9 expression through activation of Akt/ mTORC1, but not mTORC2 signaling.


Subject(s)
Cell Movement/drug effects , Matrix Metalloproteinase 9/biosynthesis , Pigment Epithelium of Eye/drug effects , Proteins/metabolism , Proto-Oncogene Proteins c-akt/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/physiology , Cells, Cultured , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Pigment Epithelium of Eye/enzymology , Pigment Epithelium of Eye/physiology , RNA Interference , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Vitreoretinopathy, Proliferative/enzymology , Vitreoretinopathy, Proliferative/pathology
9.
Eur J Cell Biol ; 91(9): 728-37, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22595285

ABSTRACT

When rhegmatogenous retinal detachment occurs, tumor necrosis factor-alpha (TNF-α) among other cytokines leaks into the subretinal space, induces resident retinal pigment epithelial (RPE) cells to migrate, which is the initial step of proliferative vitreoretinopathy (PVR). In the current study, we aim to understand how this is regulated by focusing the cellular mechanisms involved. Here we identified an Akt/Tuberous sclerosis protein 2 (TSC2)/mTOR complex1 (mTORC1) signaling pathway after TNF-α treatment to mediate RPE cell migration. Suppression of mTORC1 activation, either by its inhibitor rapamycin, or by activation of its suppressor AMP activated protein kinase (AMPK), inhibited TNF-α-mediated RPE cell migration, while RNA interference (RNAi)-mediated knocking-down of SIN1 or Rictor, two key components of mTOR complex 2 (mTORC2), had no significant effect on TNF-α-induced RPE cell migration. Our data provide initial evidence that TNF-α-mediated in vitro RPE cell migration mainly requires Akt/mTORC1, but not mTORC2 signaling. The results of this study may lead to indentify novel signaling targets against PVR.


Subject(s)
Cell Movement , Proteins/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Cell Line , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism
10.
Biochem Biophys Res Commun ; 414(3): 499-505, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21968016

ABSTRACT

Patients with age related macular degeneration (AMD) have a loss of vision in the center of the visual field. Oxidative stress plays an important role in this progress. Nerve growth factor (NGF) is important for the survival and maintenance of sympathetic and sensory neurons and NGF eye drops improve visual acuity and electro-functional activity in patients with AMD. However, the molecular mechanisms and signaling events involved in this have not been fully investigated. Using cultured human retinal pigment epithelial (RPE) cells, we demonstrate here that NGF protects RPE cells against hydrogen peroxide (H(2)O(2))-induced cell apoptosis. NGF also induces RPE cell migration, the latter is important for retinal regeneration and the recovery from AMD. H(2)O(2) decreases S6 phosphorylation and cell viability, which is restored by NGF. Rapamycin, the pharmacologic inhibitor of mammalian target of rapamycin (mTOR), diminished NGF-induced S6 phosphorylation, cell migration and protective effects against oxidative stress. Collectively, we conclude that activation of rapamycin sensitive mTOR signaling mediates NGF induced cell migration and pro-survival effects in H(2)O(2) treated RPE cells.


Subject(s)
Hydrogen Peroxide/antagonists & inhibitors , Nerve Growth Factor/pharmacology , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Retinal Pigment Epithelium/enzymology , TOR Serine-Threonine Kinases/biosynthesis , Apoptosis/drug effects , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Cytoprotection , Enzyme Activation , Humans , Hydrogen Peroxide/pharmacology , Macular Degeneration/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Retinal Pigment Epithelium/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...