Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 4 de 4
1.
Invest Ophthalmol Vis Sci ; 65(5): 3, 2024 May 01.
Article En | MEDLINE | ID: mdl-38691090

Purpose: Forty-hertz light flicker stimulation has been proven to reduce neurodegeneration, but its effect on optic nerve regeneration is unclear. This study explores the effect of 40-Hz light flicker in promoting optic nerve regeneration in zebrafish and investigates the underlying mechanisms. Methods: Wild-type and mpeg1:EGFP zebrafish were used to establish a model of optic nerve crush. Biocytin tracing and hematoxylin and eosin staining were employed to observe whether 40-Hz light flicker promotes regeneration of retinal ganglion cell axons and dendrites. Optomotor and optokinetic responses were evaluated to assess recovery of visual function. Immunofluorescence staining of mpeg1:EGFP zebrafish was performed to observe changes in microglia. Differentially expressed genes that promote optic nerve regeneration following 40-Hz light flicker stimulation were identified and validated through RNA-sequencing analysis and quantitative real-time PCR (qRT-PCR). Results: Zebrafish exhibited spontaneous optic nerve regeneration after optic nerve injury and restored visual function. We observed that 40-Hz light flicker significantly activated microglia following optic nerve injury and promoted regeneration of retinal ganglion cell axons and dendrites, as well as recovery of visual function. Transcriptomics and qRT-PCR analyses revealed that 40-Hz light flicker increased the expression of genes associated with neuronal plasticity, including bdnf, npas4a, fosab, fosb, egr4, and ier2a. Conclusions: To our knowledge, this study is the first to demonstrate that 40-Hz light flicker stimulation promotes regeneration of retinal ganglion cell axons and dendrites and recovery of visual function in zebrafish, which is associated with microglial activation and enhancement of neural plasticity.


Microglia , Nerve Regeneration , Neuronal Plasticity , Optic Nerve Injuries , Retinal Ganglion Cells , Zebrafish , Animals , Microglia/physiology , Nerve Regeneration/physiology , Optic Nerve Injuries/physiopathology , Neuronal Plasticity/physiology , Retinal Ganglion Cells/physiology , Photic Stimulation , Disease Models, Animal , Optic Nerve/physiology , Axons/physiology , Real-Time Polymerase Chain Reaction
2.
J Biophotonics ; 16(12): e202300188, 2023 12.
Article En | MEDLINE | ID: mdl-37654080

BACKGROUND: As a new technology for treating dry eye diseases, phototherapy has attracted great attention, but the research on its safety and effectiveness is limited. In this study, the therapeutic effects of low-color-temperature light-emitting diodes on dry eye in humans, rabbits, and rats were investigated. METHODS: In clinical experiments, subjects in both groups read the same paper for 3 h under light sources of two color temperatures: 1900 K (low-color-temperature light-emitting diodes) or 4000 K (artificial fluorescent white light-emitting diodes). The differences in the non-invasive tear film breakup time, tear meniscus height, and conjunctival congestion scores before and after the experiment were compared between the two groups. In animal experiments, corneal epithelial barrier function and tear production of Sprague-Dawley rats and New Zealand white rabbits with dry eye were compared before and after low-color-temperature light-emitting diodes treatment. TUNEL staining and Western blotting were used to detect the apoptosis of corneal and conjunctival cells and the expression of inflammatory factor IL-1ß. RESULTS: Low-color-temperature light-emitting diodes prolonged tear film breakup time in patients with dry eye. Moreover, it increased tear secretion, decreased fluorescein sodium staining scores, corneal and conjunctival cell apoptosis, and inflammatory factor expression in rabbits and rats with dry eye. CONCLUSIONS: Low-color-temperature light-emitting diodes phototherapy can be used as an effective treatment for dry eye, reducing its symptoms and related ocular surface damage in humans, rabbits, and rats.


Dry Eye Syndromes , Tears , Humans , Rats , Rabbits , Animals , Temperature , Tears/metabolism , Rats, Sprague-Dawley , Dry Eye Syndromes/radiotherapy , Dry Eye Syndromes/drug therapy , Conjunctiva
3.
Int Immunopharmacol ; 119: 110171, 2023 Jun.
Article En | MEDLINE | ID: mdl-37060809

Pyroptosis, an inflammasome-mediated mode of death, plays an important role in glaucoma. It has been shown that regulating the mTOR pathway can inhibit pyroptosis. Unfortunately, whether rapamycin (RAPA), a specific inhibitor of the mTOR pathway, can inhibit optic nerve crush (ONC)-induced pyroptosis to protect retinal ganglion cells (RGCs) has not been investigated. Our research aimed to confirm the effect of intravitreal injection of RAPA on RGCs. Furthermore, we used the ONC model to explore the underlying mechanisms. First, we observed that intravitreal injection of RAPA alleviated RGC damage induced by various types of injury. We then used the ONC model to further explore the potential mechanism of RAPA. Mechanistically, RAPA not only reduced the activation of glial cells in the retina but also inhibited retinal pyroptosis-induced expression of inflammatory factors such as nucleotide-binding oligomeric domain-like receptor 3 (NLRP3), apoptosis-associated speckle-like protein containing a CARD (ASC), N-terminal of gasdermin-D (GSDMD-N), IL-18 and IL-1ß. Moreover, RAPA exerted protective effects on RGC axons, possibly by inhibiting glial activation and regulating the mTOR/ROCK pathway. Therefore, this study demonstrates a novel mechanism by which RAPA protects against glaucoma and provides further evidence for its application in preclinical studies.


Glaucoma , Optic Nerve Injuries , Humans , Animals , Retinal Ganglion Cells , Sirolimus/pharmacology , Sirolimus/therapeutic use , Neuroinflammatory Diseases , Optic Nerve Injuries/drug therapy , Optic Nerve , TOR Serine-Threonine Kinases/metabolism , Glaucoma/drug therapy , Disease Models, Animal
4.
Cancer Gene Ther ; 26(7-8): 195-207, 2019 07.
Article En | MEDLINE | ID: mdl-30470842

Understanding the molecular mechanisms for the development of non-Hodgkin lymphoma (NHL) will improve our ability to cure the patients. qRT-PCR was applied for the examination of the efficiency of shRNA for DNMT1, the expression of suppressor genes, miRNA-152. The MTT analysis, cell cycle analysis, clonal formation, and apoptotic analysis were used to examine the functions of DNMT1 and miR-152 in lymphoma cells. Methylation-specific polymerase chain reaction (MSP) was used to examine the methylation of tumor suppressor genes. The dual luciferase assay and western blot were used to validate if DNMT1 is the target of miR-152. For the in vivo experiments, the lymphoma cells were injected into the nude mice for quantification of the tumor growth after transfection of miR-152 mimics. Knockdown of DNMT1 by shRNA (sh-DNMT1) in OCI-Ly10 and Granta-159 cells significantly upregulated the expression of tumor suppressor genes (SOCS3, BCL2L10, p16, p14, and SHP-1) via decreasing their methylation level. At the cellular level, we found sh-DNMT1 inhibited the proliferation, clonal formation and cell cycle progression and induced the cell apoptosis of lymphoma cells. Furthermore, we found miR-152 can downregulates the expression of DNMT1 via directly targeting the gene. Overexpression of miR-152 also increased the expression of tumor suppressor genes SOCS3 and SHP-1. And miR-152 also can inhibit the cell proliferation and induce the cell apoptosis. Moreover, we found overexpression of miR-152 significantly repressed the tumor growth with decreased DNMT1 expression and increased expression of tumor suppressor genes in vivo. Our study demonstrates that miR-152 can inhibit lymphoma growth via suppressing DNMT1-mediated silencing of SOCS3 and SHP-1. These data demonstrate a new mechanism for the development of NHL and this may provide a new therapeutic target for NHL.


DNA (Cytosine-5-)-Methyltransferase 1/genetics , Lymphoma, Non-Hodgkin/genetics , MicroRNAs/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Down-Regulation , Gene Knockdown Techniques , Genes, Tumor Suppressor , HEK293 Cells , Heterografts , Humans , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Mice , MicroRNAs/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transfection
...