Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.826
Filtrer
1.
Commun Biol ; 7(1): 807, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961177

RÉSUMÉ

Glaucoma is the leading cause of irreversible blindness and is characterized by progressive retinal ganglion cell (RGC) loss and retinal nerve fiber layer thinning. Currently, no existing treatment is effective for the preservation of RGCs. MicroRNA-22-3p (miR22) and small extracellular vesicles derived from mesenchymal stem cells (MSC-sEVs) have neuroprotective effects. In this study, we apply miR22-overexpressing MSC-sEVs in an N-methyl-D-aspartic acid (NMDA)-induced RGC injury model to assess their short-term therapeutic effects and explore the underlying mechanisms. We find that mice in the miR22-sEVs-treated group have thicker retinas, fewer apoptotic cells, more reserved RGCs, better retinal function, and lower expression levels of Bax and caspase-3. MiR22-sEVs treatment promotes viability, inhibits apoptosis and inhibits Bax and caspase-3 expression in RGC-5 cells. MiR22 targets mitogen-activated protein kinase kinase kinase 12 to inhibit apoptosis by regulating the mitogen-activated protein kinase (MAPK) signaling pathway. Collectively, our results suggest that miR22-sEVs ameliorate NMDA-induced RGC injury through the inhibition of MAPK signaling pathway-mediated apoptosis, providing a potential therapy for glaucoma and other diseases that involve RGC damage.


Sujet(s)
Vésicules extracellulaires , Système de signalisation des MAP kinases , Cellules souches mésenchymateuses , microARN , Cellules ganglionnaires rétiniennes , Cellules ganglionnaires rétiniennes/métabolisme , microARN/génétique , microARN/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Vésicules extracellulaires/métabolisme , Souris , Apoptose , Souris de lignée C57BL , Glaucome/génétique , Glaucome/métabolisme , Glaucome/anatomopathologie , Glaucome/thérapie , Mâle
2.
Sci Rep ; 14(1): 14907, 2024 06 28.
Article de Anglais | MEDLINE | ID: mdl-38942959

RÉSUMÉ

To evaluate the protective effect of gallic acid on the optic nerve by studying the inhibitory effect of gallic acid on oxidative stress in retinal ganglion cells. 100 male SD rats were randomly divided into four groups: normal control group, simple high IOP group, 0.5% gallic acid experimental group, and 1% gallic acid experimental group. HE staining, immunofluorescence, DHE staining, Western blot, and q-PCR were used to observe the antioxidant effect of gallic acid on the retina of acute ocular hypertension rats. HE staining of the retina of SD rats confirmed that the nucleus of RGCs was clear, the thickness of the RNFL was regular in the normal control group, and the nucleus of RGCs was ruptured and lysed in the simple high intraocular pressure (IOP) group and the gallic acid group, and the thickness of the RNFL was significantly thickened, but the thickness of the RNFL in the gallic acid group was significantly reduced compared with that in the simple high IOP group (p < 0.05). DHE staining showed that ROS content in the simple high IOP group was significantly increased compared with the normal control group, and ROS content was significantly decreased after the application of gallic acid (p < 0.05). Immunofluorescence staining with Brn-3a antibody confirmed that the number of RGCs was significantly reduced in the simple high IOP group compared with the normal control group, whereas after application of gallic acid, the number of RGCs was significantly more in the gallic acid group than in the simple high IOP group (p < 0.05). Western Blot and q-PCR confirmed that hypoxia-inducing factor 1α (HIF-1α) protein content and transcription level were significantly increased in the retinal tissue of the simple high IOP group, and gallic acid could inhibit HIF-1α protein content (p < 0.05) and reduce transcription factor level (p < 0.05). Gallic acid exerts a protective effect on RGC by inhibiting oxidative stress in rats with acute IOP elevation.


Sujet(s)
Antioxydants , Modèles animaux de maladie humaine , Acide gallique , Glaucome , Stress oxydatif , Rat Sprague-Dawley , Cellules ganglionnaires rétiniennes , Acide gallique/pharmacologie , Animaux , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Antioxydants/pharmacologie , Mâle , Rats , Glaucome/métabolisme , Glaucome/traitement médicamenteux , Glaucome/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Pression intraoculaire/effets des médicaments et des substances chimiques , Hypertension oculaire/traitement médicamenteux , Hypertension oculaire/métabolisme , Hypertension oculaire/anatomopathologie
3.
Cells ; 13(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38920659

RÉSUMÉ

Recent emerging studies have demonstrated numerous critical roles of exosomes in cell-to-cell signaling. We investigated exosomes in the aqueous humor of glaucoma patients and controls and compared their characteristics with other biomarkers such as cytokines. Glaucoma patients exhibited higher exosome particle counts and smaller sizes compared to controls. Higher exosome density was correlated with more severe visual field loss. Conversely, concentrations of aqueous humor cytokines, particularly PD-L1, were primarily associated with intraocular pressure, and none of the cytokines showed a significant association with visual field damage. This may reflect the characteristics of exosomes, which are advantageous for crossing various biological barriers. Exosomes may contain more information about glaucoma functional damage occurring in the retina or optic nerve head. This highlights the potential importance of exosomes as signaling mediators distinct from other existing molecules.


Sujet(s)
Humeur aqueuse , Marqueurs biologiques , Cytokines , Exosomes , Glaucome , Humains , Humeur aqueuse/métabolisme , Exosomes/métabolisme , Marqueurs biologiques/métabolisme , Glaucome/métabolisme , Glaucome/anatomopathologie , Cytokines/métabolisme , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Pression intraoculaire , Études cas-témoins
4.
Cells ; 13(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38920673

RÉSUMÉ

In the context of glaucoma, intraocular pressure (IOP) and age are recognized as the primary factors contributing to its onset and progression. However, significant reductions in IOP fail to completely halt its advancement. An emerging body of literature highlights the role of neuroinflammation in glaucoma. This study aimed to explore Bromfenac's anti-inflammatory properties in mitigating neuroinflammation associated with glaucoma using an ischemia-reperfusion (IR) glaucoma model. Bromfenac's impact on microglia and astrocytes under pressure was assessed via Western blotting and an enzyme-linked immunosorbent assay. Immunohistochemical staining was used to evaluate glial activation and changes in inflammatory marker expression in the IR model. Bromfenac led to the downregulation of inflammatory markers, which were elevated in the conditions of elevated pressure, and necroptosis markers were downregulated in astrocytes. In the IR model, elevated levels of GFAP and Iba-1 indicated glial activation. Following Bromfenac administration, levels of iNOS, COX-2, and PGE2-R were reduced, suggesting a decrease in neuroinflammation. Furthermore, Bromfenac administration in the IR model resulted in the improved survival of retinal ganglion cells (RGCs) and preservation of retinal function, as demonstrated by immunohistochemical staining and electroretinography. In summary, Bromfenac proved effective in diminishing neuroinflammation and resulted in enhanced RGC survival.


Sujet(s)
Astrocytes , Benzophénones , Bromobenzènes , Modèles animaux de maladie humaine , Glaucome , Lésion d'ischémie-reperfusion , Bromobenzènes/pharmacologie , Bromobenzènes/usage thérapeutique , Animaux , Benzophénones/pharmacologie , Benzophénones/usage thérapeutique , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/complications , Glaucome/traitement médicamenteux , Glaucome/anatomopathologie , Glaucome/complications , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/anatomopathologie , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Cellules ganglionnaires rétiniennes/anatomopathologie , Cellules ganglionnaires rétiniennes/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Microglie/anatomopathologie , Mâle , Pression intraoculaire/effets des médicaments et des substances chimiques , Rats
5.
PLoS One ; 19(6): e0305624, 2024.
Article de Anglais | MEDLINE | ID: mdl-38889111

RÉSUMÉ

OBJECTIVE: To investigate the relationship between corneal diameter and internal corneal span determined from angle-to-angle distance using ultrasound biomicroscopy (UBM) in an observational cross-sectional patient population comprised of 54 eyes (28 healthy control eyes, ages 0.1 to 11.3 years; 26 eyes with primary congenital glaucoma, ages 0.1 to 3.5 years) from 41 pediatric participants ages 0.1 to 11.3 years (mean age: 3±3 years, median age: 2 years). METHODS: Forty cornea photographs with reference ruler and 110 UBM images were obtained. Three observers measured horizontal and vertical corneal diameter and angle-to-angle distance in each cornea photo and UBM image using ImageJ and the average values were used. Main outcome measures were Pearson correlation coefficient, linear regression, mean difference between corneal diameter and angle-to-angle distance, and intra-class correlation coefficients among measurements from all three observers for each parameter. RESULTS: Corneal diameter and angle-to-angle distance had a strong positive correlation horizontally (Pearson r = 0.89, p<0.001) and vertically (r = 0.93, p<0.001). Correlation was consistent regardless of presence of primary congenital glaucoma and participant age. Regression analysis demonstrated a linear relationship between the parameters for horizontal (CD = 0.99*AA+0.28, R2 = 0.81, p<0.001) and vertical (CD = 0.91 *AA+1.32, R2 = 0.85, p<0.001) dimensions. Overall, reliability was good-excellent, ranging from an ICC of 0.76 for vertical corneal diameter to 0.90 for horizontal angle-to-angle distance. CONCLUSIONS: Based on the strong positive correlation found between corneal diameter and angle-to-angle distance in our study population, UBM image analysis can be used to accurately estimate corneal diameter from angle-to-angle distance in children with healthy eyes and primary congenital glaucoma. UBM may provide a useful intraocular alternative for estimating corneal diameter and monitoring diseases that affect the cornea in infants and children, such as congenital glaucoma.


Sujet(s)
Cornée , Microscopie acoustique , Humains , Enfant d'âge préscolaire , Cornée/imagerie diagnostique , Enfant , Microscopie acoustique/méthodes , Mâle , Femelle , Nourrisson , Études transversales , Glaucome/imagerie diagnostique , Glaucome/anatomopathologie
6.
Transl Vis Sci Technol ; 13(6): 10, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38884547

RÉSUMÉ

Purpose: To explore the structural-functional loss relationship from optic-nerve-head- and macula-centred spectral-domain (SD) Optical Coherence Tomography (OCT) images in the full spectrum of glaucoma patients using deep-learning methods. Methods: A cohort comprising 5238 unique eyes classified as suspects or diagnosed with glaucoma was considered. All patients underwent ophthalmologic examination consisting of standard automated perimetry (SAP), macular OCT, and peri-papillary OCT on the same day. Deep learning models were trained to estimate G-pattern visual field (VF) mean deviation (MD) and cluster MD using retinal thickness maps from seven layers: retinal nerve fiber layer (RNFL), ganglion cell layer and inner plexiform layer (GCL + IPL), inner nuclear layer and outer plexiform layer (INL + OPL), outer nuclear layer (ONL), photoreceptors and retinal pigmented epithelium (PR + RPE), choriocapillaris and choroidal stroma (CC + CS), total retinal thickness (RT). Results: The best performance on MD prediction is achieved by RNFL, GCL + IPL and RT layers, with R2 scores of 0.37, 0.33, and 0.31, respectively. Combining macular and peri-papillary scans outperforms single modality prediction, achieving an R2 value of 0.48. Cluster MD predictions show promising results, notably in central clusters, reaching an R2 of 0.56. Conclusions: The combination of multiple modalities, such as optic-nerve-head circular B-scans and retinal thickness maps from macular SD-OCT images, improves the performance of MD and cluster MD prediction. Our proposed model demonstrates the highest level of accuracy in predicting MD in the early-to-mid stages of glaucoma. Translational Relevance: Objective measures recorded with SD-OCT can optimize the number of visual field tests and improve individualized glaucoma care by adjusting VF testing frequency based on deep-learning estimates of functional damage.


Sujet(s)
Apprentissage profond , Macula , Tomographie par cohérence optique , Champs visuels , Tomographie par cohérence optique/méthodes , Humains , Femelle , Adulte d'âge moyen , Mâle , Champs visuels/physiologie , Macula/imagerie diagnostique , Macula/anatomopathologie , Pronostic , Sujet âgé , Cellules ganglionnaires rétiniennes/anatomopathologie , Glaucome/imagerie diagnostique , Glaucome/anatomopathologie , Neurofibres/anatomopathologie , Tests du champ visuel/méthodes , Papille optique/imagerie diagnostique , Papille optique/anatomopathologie
7.
J Neuroinflammation ; 21(1): 145, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824526

RÉSUMÉ

BACKGROUND: Recent experimental studies of neuroinflammation in glaucoma pointed to cFLIP as a molecular switch for cell fate decisions, mainly regulating cell type-specific caspase-8 functions in cell death and inflammation. This study aimed to determine the importance of cFLIP for regulating astroglia-driven neuroinflammation in experimental glaucoma by analyzing the outcomes of astroglia-targeted transgenic deletion of cFLIP or cFLIPL. METHODS: Glaucoma was modeled by anterior chamber microbead injections to induce ocular hypertension in mouse lines with or without conditional deletion of cFLIP or cFLIPL in astroglia. Morphological analysis of astroglia responses assessed quantitative parameters in retinal whole mounts immunolabeled for GFAP and inflammatory molecules or assayed for TUNEL. The molecular analysis included 36-plexed immunoassays of the retina and optic nerve cytokines and chemokines, NanoString-based profiling of inflammation-related gene expression, and Western blot analysis of selected proteins in freshly isolated samples of astroglia. RESULTS: Immunoassays and immunolabeling of retina and optic nerve tissues presented reduced production of various proinflammatory cytokines, including TNFα, in GFAP/cFLIP and GFAP/cFLIPL relative to controls at 12 weeks of ocular hypertension with no detectable alteration in TUNEL. Besides presenting a similar trend of the proinflammatory versus anti-inflammatory molecules displayed by immunoassays, NanoString-based molecular profiling detected downregulated NF-κB/RelA and upregulated RelB expression of astroglia in ocular hypertensive samples of GFAP/cFLIP compared to ocular hypertensive controls. Analysis of protein expression also revealed decreased phospho-RelA and increased phospho-RelB in parallel with an increase in caspase-8 cleavage products. CONCLUSIONS: A prominent response limiting neuroinflammation in ocular hypertensive eyes with cFLIP-deletion in astroglia values the role of cFLIP in the molecular regulation of glia-driven neuroinflammation during glaucomatous neurodegeneration. The molecular responses accompanying the lessening of neurodegenerative inflammation also seem to maintain astroglia survival despite increased caspase-8 cleavage with cFLIP deletion. A transcriptional autoregulatory response, dampening RelA but boosting RelB for selective expression of NF-κB target genes, might reinforce cell survival in cFLIP-deleted astroglia.


Sujet(s)
Astrocytes , Protéine de régulation de l'apoptose CASP8 et FADD-like , Glaucome , Maladies neuro-inflammatoires , Animaux , Protéine de régulation de l'apoptose CASP8 et FADD-like/métabolisme , Protéine de régulation de l'apoptose CASP8 et FADD-like/génétique , Souris , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Glaucome/métabolisme , Glaucome/anatomopathologie , Glaucome/génétique , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Souris transgéniques , Modèles animaux de maladie humaine , Cytokines/métabolisme , Rétine/métabolisme , Rétine/anatomopathologie , Souris de lignée C57BL , Nerf optique/anatomopathologie , Nerf optique/métabolisme , Protéine gliofibrillaire acide/métabolisme
8.
Acta Neuropathol Commun ; 12(1): 89, 2024 06 07.
Article de Anglais | MEDLINE | ID: mdl-38845058

RÉSUMÉ

The microtubule-associated protein Tau is a key player in various neurodegenerative conditions, including Alzheimer's disease (AD) and Tauopathies, where its hyperphosphorylation disrupts neuronal microtubular lattice stability. Glaucoma, a neurodegenerative disorder affecting the retina, leads to irreversible vision loss by damaging retinal ganglion cells and the optic nerve, often associated with increased intraocular pressure. Prior studies have indicated Tau expression and phosphorylation alterations in the retina in both AD and glaucoma, yet the causative or downstream nature of Tau protein changes in these pathologies remains unclear. This study investigates the impact of Tau protein modulation on retinal neurons under normal and experimental glaucoma conditions. Employing AAV9-mediated gene therapy for Tau overexpression and knockdown, both manipulations were found to adversely affect retinal structural and functional measures as well as neuroprotective Akt/Erk survival signalling in healthy conditions. In the experimental glaucoma model, Tau overexpression intensified inner retinal degeneration, while Tau silencing provided significant protection against these degenerative changes. These findings underscore the critical role of endogenous Tau protein levels in preserving retinal integrity and emphasize the therapeutic potential of targeting Tau in glaucoma pathology.


Sujet(s)
Thérapie génétique , Glaucome , Protéines tau , Protéines tau/métabolisme , Animaux , Glaucome/métabolisme , Glaucome/anatomopathologie , Glaucome/génétique , Thérapie génétique/méthodes , Protéines proto-oncogènes c-akt/métabolisme , Dependovirus/génétique , Modèles animaux de maladie humaine , Dégénérescence de la rétine/métabolisme , Dégénérescence de la rétine/anatomopathologie , Dégénérescence de la rétine/génétique , Rétine/métabolisme , Rétine/anatomopathologie , Système de signalisation des MAP kinases/physiologie , Transduction du signal/physiologie , Souris , Souris de lignée C57BL , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Phénotype
9.
Int J Mol Sci ; 25(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891923

RÉSUMÉ

The ocular glymphatic system subserves the bidirectional polarized fluid transport in the optic nerve, whereby cerebrospinal fluid from the brain is directed along periarterial spaces towards the eye, and fluid from the retina is directed along perivenous spaces following upon its axonal transport across the glial lamina. Fluid homeostasis and waste removal are vital for retinal function, making the ocular glymphatic fluid pathway a potential route for targeted manipulation to combat blinding ocular diseases such as age-related macular degeneration, diabetic retinopathy, and glaucoma. Several lines of work investigating the bidirectional ocular glymphatic transport with varying methodologies have developed diverging mechanistic models, which has created some confusion about how ocular glymphatic transport should be defined. In this review, we provide a comprehensive summary of the current understanding of the ocular glymphatic system, aiming to address misconceptions and foster a cohesive understanding of the topic.


Sujet(s)
Système glymphatique , Humains , Système glymphatique/physiologie , Système glymphatique/métabolisme , Animaux , Nerf optique/métabolisme , Nerf optique/physiologie , Rétine/métabolisme , Rétine/physiologie , Oeil/métabolisme , Glaucome/métabolisme , Glaucome/physiopathologie , Glaucome/anatomopathologie
10.
Exp Cell Res ; 440(1): 114137, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38897410

RÉSUMÉ

Glaucoma is characterized by pathological elevation of intraocular pressure (IOP) due to dysfunctional trabecular meshwork (TM), which is the primary cause of irreversible vision loss. There are currently no effective treatment strategies for glaucoma. Mitochondrial function plays a crucial role in regulating IOP within the TM. In this study, primary TM cells treated with dexamethasone were used to simulate glaucomatous changes, showing abnormal cellular cytoskeleton, increased expression of extracellular matrix, and disrupted mitochondrial fusion and fission dynamics. Furthermore, glaucomatous TM cell line GTM3 exhibited impaired mitochondrial membrane potential and phagocytic function, accompanied by decreased oxidative respiratory levels as compared to normal TM cells iHTM. Mechanistically, lower NAD + levels in GTM3, possibly associated with increased expression of key enzymes CD38 and PARP1 related to NAD + consumption, were observed. Supplementation of NAD + restored mitochondrial function and cellular viability in GTM3 cells. Therefore, we propose that the aberrant mitochondrial function in glaucomatous TM cells may be attributed to increased NAD + consumption dependent on CD38 and PARP1, and NAD + supplementation could effectively ameliorate mitochondrial function and improve TM function, providing a novel alternative approach for glaucoma treatment.


Sujet(s)
Glaucome , Mitochondries , NAD , Réseau trabéculaire de la sclère , Réseau trabéculaire de la sclère/métabolisme , Réseau trabéculaire de la sclère/effets des médicaments et des substances chimiques , Réseau trabéculaire de la sclère/anatomopathologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/anatomopathologie , Glaucome/métabolisme , Glaucome/anatomopathologie , Glaucome/traitement médicamenteux , NAD/métabolisme , Humains , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Pression intraoculaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Antigènes CD38/métabolisme , Antigènes CD38/génétique , Lignée cellulaire , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , Dexaméthasone/pharmacologie , Cellules cultivées
11.
Cell Rep Med ; 5(5): 101554, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38729157

RÉSUMÉ

The axons of retinal ganglion cells (RGCs) form the optic nerve, transmitting visual information from the eye to the brain. Damage or loss of RGCs and their axons is the leading cause of visual functional defects in traumatic injury and degenerative diseases such as glaucoma. However, there are no effective clinical treatments for nerve damage in these neurodegenerative diseases. Here, we report that LIM homeodomain transcription factor Lhx2 promotes RGC survival and axon regeneration in multiple animal models mimicking glaucoma disease. Furthermore, following N-methyl-D-aspartate (NMDA)-induced excitotoxicity damage of RGCs, Lhx2 mitigates the loss of visual signal transduction. Mechanistic analysis revealed that overexpression of Lhx2 supports axon regeneration by systematically regulating the transcription of regeneration-related genes and inhibiting transcription of Semaphorin 3C (Sema3C). Collectively, our studies identify a critical role of Lhx2 in promoting RGC survival and axon regeneration, providing a promising neural repair strategy for glaucomatous neurodegeneration.


Sujet(s)
Axones , Modèles animaux de maladie humaine , Glaucome , Protéines à homéodomaine LIM , Régénération nerveuse , Cellules ganglionnaires rétiniennes , Facteurs de transcription , Animaux , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Protéines à homéodomaine LIM/métabolisme , Protéines à homéodomaine LIM/génétique , Glaucome/génétique , Glaucome/anatomopathologie , Glaucome/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Axones/métabolisme , Axones/anatomopathologie , Souris , Régénération nerveuse/génétique , Régénération nerveuse/physiologie , Souris de lignée C57BL , Survie cellulaire/génétique , Sémaphorines/métabolisme , Sémaphorines/génétique , N-Méthyl-aspartate/métabolisme
12.
Biomed Pharmacother ; 175: 116711, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38735082

RÉSUMÉ

Glaucoma, the leading cause of irreversible blindness worldwide, is characterized by neurodegeneration and neuroinflammation with retinal NAD/NADP and GSH decline. Nicotinamide adenine dinucleotide (NAD)/NAD phosphate (NADP) and glutathione (GSH) are two redox reducers in neuronal and glial metabolism. However, therapeutic strategies targeting NAD/NADP or GSH do not exert ideal effects, and the underlying mechanisms are still poorly understood. We assessed morphological changes in retinal ganglion cells (RGCs), the affected neurons in glaucoma, and Müller cells, the major glial cells in the retina, as well as the levels of phosphorylated p38 (p-p38) and Caspase-3 in glaucoma patients. We constructed a modified chronic ocular hypertensive rat model and an oxygen-glucose deprivation (OGD) cell model. After applying NADPH and N-acetylcysteine (NAC), a precursor to cysteine, the rate-limiting substrate in GSH biosynthesis, to cells, apoptosis, axonal damage and peroxidation were reduced in the RGCs of the NAC group and p-p38 levels were decreased in the RGCs of the NADPH group, while in stimulated Müller cells cultured individually or cocultured with RGCs, gliosis and p38/MAPK, rather than JNK/MAPK, activation were inhibited. The results were more synergistic in the rat model, where either NADPH or NAC showed crossover effects on inhibiting peroxidation and p38/MAPK pathway activation. Moreover, the combination of NADPH and NAC ameliorated RGC electrophysiological function and prevented Müller cell gliosis to the greatest extent. These data illustrated conjoined mechanisms in glaucomatous RGC injury and Müller cell gliosis and suggested that NADPH and NAC collaborate as a neuroprotective and anti-inflammatory combination treatment for glaucoma and other underlying human neurodegenerative diseases.


Sujet(s)
Acétylcystéine , NADP , Hypertension oculaire , Rat Sprague-Dawley , Cellules ganglionnaires rétiniennes , p38 Mitogen-Activated Protein Kinases , Animaux , NADP/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , Hypertension oculaire/métabolisme , Hypertension oculaire/traitement médicamenteux , Hypertension oculaire/anatomopathologie , Acétylcystéine/pharmacologie , Rats , Mâle , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Glaucome/métabolisme , Glaucome/anatomopathologie , Glaucome/traitement médicamenteux , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme , Humains , Cellules épendymogliales/effets des médicaments et des substances chimiques , Cellules épendymogliales/métabolisme , Cellules épendymogliales/anatomopathologie , Modèles animaux de maladie humaine , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Maladie chronique , Neuroprotecteurs/pharmacologie , Cellules cultivées , Peroxydation lipidique/effets des médicaments et des substances chimiques
13.
Exp Eye Res ; 244: 109917, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38697276

RÉSUMÉ

In glaucoma, scleral fibroblasts are exposed to IOP-associated mechanical strain and elevated TGFß levels. These stimuli, in turn, lead to scleral remodeling. Here, we examine the scleral fibroblast migratory and transcriptional response to these stimuli to better understand mechanisms of glaucomatous scleral remodeling. Human peripapillary scleral (PPS) fibroblasts were cultured on parallel grooves, treated with TGFß (2 ng/ml) in the presence of vehicle or TGFß signaling inhibitors, and exposed to uniaxial strain (1 Hz, 5%, 12-24 h). Axis of cellular orientation was determined at baseline, immediately following strain, and 24 h after strain cessation with 0° being completely aligned with grooves and 90° being perpendicular. Fibroblasts migration in-line and across grooves was assessed using a scratch assay. Transcriptional profiling of TGFß-treated fibroblasts with or without strain was performed by RT-qPCR and pERK, pSMAD2, and pSMAD3 levels were measured by immunoblot. Pre-strain alignment of TGFß-treated cells with grooves (6.2 ± 1.5°) was reduced after strain (21.7 ± 5.3°, p < 0.0001) and restored 24 h after strain cessation (9.5 ± 2.6°). ERK, FAK, and ALK5 inhibition prevented this reduction; however, ROCK, YAP, or SMAD3 inhibition did not. TGFß-induced myofibroblast markers were reduced by strain (αSMA, POSTN, ASPN, MLCK1). While TGFß-induced phosphorylation of ERK and SMAD2 was unaffected by cyclic strain, SMAD3 phosphorylation was reduced (p = 0.0004). Wound healing across grooves was enhanced by ROCK and SMAD3 inhibition but not ERK or ALK5 inhibition. These results provide insight into the mechanisms by which mechanical strain alters the cellular response to TGFß and the potential signaling pathways that underlie scleral remodeling.


Sujet(s)
Mouvement cellulaire , Fibroblastes , Sclère , Contrainte mécanique , Facteur de croissance transformant bêta , Humains , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Cellules cultivées , Facteur de croissance transformant bêta/pharmacologie , Facteur de croissance transformant bêta/métabolisme , Sclère/métabolisme , Transduction du signal , Réaction de polymérisation en chaine en temps réel , Régulation de l'expression des gènes , Glaucome/métabolisme , Glaucome/anatomopathologie
14.
Invest Ophthalmol Vis Sci ; 65(5): 15, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38717426

RÉSUMÉ

Purpose: Mutations in the genes encoding type IV collagen alpha 1 (COL4A1) and alpha 2 (COL4A2) cause a multisystem disorder that includes ocular anterior segment dysgenesis (ASD) and glaucoma. We previously showed that transforming growth factor beta (TGFß) signaling was elevated in developing anterior segments from Col4a1 mutant mice and that reducing TGFß signaling ameliorated ASD, supporting a role for the TGFß pathway in disease pathogenesis. Here, we tested whether altered TGFß signaling also contributes to glaucoma-related phenotypes in Col4a1 mutant mice. Methods: To test the role of TGFß signaling in glaucoma-relevant phenotypes, we genetically reduced TGFß signaling using mice with mutated Tgfbr2, which encodes the common receptor for all TGFß ligands in Col4a1+/G1344D mice. We performed slit-lamp biomicroscopy and optical coherence tomography for qualitative and quantitative analyses of anterior and posterior ocular segments, histological analyses of ocular tissues and optic nerves, and intraocular pressure assessments using rebound tonometry. Results: Col4a1+/G1344D mice showed defects of the ocular drainage structures, including iridocorneal adhesions, and phenotypes consistent with glaucomatous neurodegeneration, including thinning of the nerve fiber layer, retinal ganglion cell loss, optic nerve head excavation, and optic nerve degeneration. We found that reducing TGFß receptor 2 (TGFBR2) was protective for ASD, ameliorated ocular drainage structure defects, and protected against glaucomatous neurodegeneration in Col4a1+/G1344D mice. Conclusions: Our results suggest that elevated TGFß signaling contributes to glaucomatous neurodegeneration in Col4a1 mutant mice.


Sujet(s)
Collagène de type IV , Glaucome , Récepteur de type II du facteur de croissance transformant bêta , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Souris , Pôle antérieur du bulbe oculaire/métabolisme , Pôle antérieur du bulbe oculaire/anatomopathologie , Collagène de type IV/métabolisme , Collagène de type IV/génétique , Modèles animaux de maladie humaine , Glaucome/métabolisme , Glaucome/génétique , Glaucome/anatomopathologie , Pression intraoculaire/physiologie , Souris de lignée C57BL , Mutation , Nerf optique/anatomopathologie , Nerf optique/métabolisme , Atteintes du nerf optique/métabolisme , Atteintes du nerf optique/génétique , Phénotype , Récepteur de type II du facteur de croissance transformant bêta/génétique , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Cellules ganglionnaires rétiniennes/métabolisme , Transduction du signal/physiologie , Biomicroscopie , Tomographie par cohérence optique , Tonométrie oculaire , Facteur de croissance transformant bêta/métabolisme
15.
FASEB J ; 38(10): e23651, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38752537

RÉSUMÉ

Singleton-Merten syndrome (SMS) is a rare immunogenetic disorder affecting multiple systems, characterized by dental dysplasia, aortic calcification, glaucoma, skeletal abnormalities, and psoriasis. Glaucoma, a key feature of both classical and atypical SMS, remains poorly understood in terms of its molecular mechanism caused by DDX58 mutation. This study presented a novel DDX58 variant (c.1649A>C [p.Asp550Ala]) in a family with childhood glaucoma. Functional analysis showed that DDX58 variant caused an increase in IFN-stimulated gene expression and high IFN-ß-based type-I IFN. As the trabecular meshwork (TM) is responsible for controlling intraocular pressure (IOP), we examine the effect of IFN-ß on TM cells. Our study is the first to demonstrate that IFN-ß significantly reduced TM cell viability and function by activating autophagy. In addition, anterior chamber injection of IFN-ß remarkably increased IOP level in mice, which can be attenuated by treatments with autophagy inhibitor chloroquine. To uncover the specific mechanism underlying IFN-ß-induced autophagy in TM cells, we performed microarray analysis in IFN-ß-treated and DDX58 p.Asp550Ala TM cells. It showed that RSAD2 is necessary for IFN-ß-induced autophagy. Knockdown of RSAD2 by siRNA significantly decreased autophagy flux induced by IFN-ß. Our findings suggest that DDX58 mutation leads to the overproduction of IFN-ß, which elevates IOP by modulating autophagy through RSAD2 in TM cells.


Sujet(s)
Autophagie , Interféron bêta , Pression intraoculaire , Réseau trabéculaire de la sclère , Autophagie/effets des médicaments et des substances chimiques , Réseau trabéculaire de la sclère/métabolisme , Réseau trabéculaire de la sclère/effets des médicaments et des substances chimiques , Humains , Animaux , Souris , Pression intraoculaire/physiologie , Interféron bêta/métabolisme , Mâle , Femelle , Glaucome/anatomopathologie , Glaucome/métabolisme , Glaucome/génétique , Surdité neurosensorielle/génétique , Surdité neurosensorielle/anatomopathologie , Surdité neurosensorielle/métabolisme , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Souris de lignée C57BL , Mutation , Atrophie optique/génétique , Atrophie optique/métabolisme , Atrophie optique/anatomopathologie , Pedigree , Odontodysplasie , Calcification vasculaire , Hypoplasie de l'émail dentaire , Métacarpe/malformations , Ostéoporose , Maladies musculaires , Maladies de l'aorte , Récepteurs immunologiques
16.
J Affect Disord ; 358: 487-499, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38705527

RÉSUMÉ

BACKGROUND: Glaucoma, a progressive neurodegenerative disorder leading to irreversible blindness, is associated with heightened rates of generalized anxiety and depression. This study aims to comprehensively investigate brain morphological changes in glaucoma patients, extending beyond visual processing areas, and explores overlaps with morphological alterations observed in anxiety and depression. METHODS: A comparative meta-analysis was conducted, using case-control studies of brain structural integrity in glaucoma patients. We aimed to identify regions with gray matter volume (GMV) changes, examine their role within distinct large-scale networks, and assess overlap with alterations in generalized anxiety disorder (GAD) and major depressive disorder (MDD). RESULTS: Glaucoma patients exhibited significant GMV reductions in visual processing regions (lingual gyrus, thalamus). Notably, volumetric reductions extended beyond visual systems, encompassing the left putamen and insula. Behavioral and functional network decoding revealed distinct large-scale networks, implicating visual, motivational, and affective domains. The insular region, linked to pain and affective processes, displayed reductions overlapping with alterations observed in GAD. LIMITATIONS: While the study identified significant morphological alterations, the number of studies from both the glaucoma and GAD cohorts remains limited due to the lack of independent studies meeting our inclusion criteria. CONCLUSION: The study proposes a tripartite brain model for glaucoma, with visual processing changes related to the lingual gyrus and additional alterations in the putamen and insular regions tied to emotional or motivational functions. These neuroanatomical changes extend beyond the visual system, implying broader implications for brain structure and potential pathological developments, providing insights into the overall neurological consequences of glaucoma.


Sujet(s)
Troubles anxieux , Trouble dépressif majeur , Glaucome , Substance grise , Humains , Glaucome/anatomopathologie , Glaucome/physiopathologie , Substance grise/anatomopathologie , Substance grise/imagerie diagnostique , Troubles anxieux/anatomopathologie , Troubles anxieux/imagerie diagnostique , Trouble dépressif majeur/anatomopathologie , Trouble dépressif majeur/imagerie diagnostique , Imagerie par résonance magnétique , Encéphale/anatomopathologie , Encéphale/imagerie diagnostique , Régulation émotionnelle/physiologie , Études cas-témoins , Putamen/anatomopathologie , Putamen/imagerie diagnostique
17.
J Transl Med ; 22(1): 440, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38720358

RÉSUMÉ

PURPOSE: To explore the impact of microRNA 146a (miR-146a) and the underlying mechanisms in profibrotic changes following glaucoma filtering surgery (GFS) in rats and stimulation by transforming growth factor (TGF)-ß1 in rat Tenon's capsule fibroblasts. METHODS: Cultured rat Tenon's capsule fibroblasts were treated with TGF-ß1 and analyzed with microarrays for mRNA profiling to validate miR-146a as the target. The Tenon's capsule fibroblasts were then respectively treated with lentivirus-mediated transfection of miR-146a mimic or inhibitor following TGF-ß1 stimulation in vitro, while GFS was performed in rat eyes with respective intraoperative administration of miR-146a, mitomycin C (MMC), or 5-fluorouracil (5-FU) in vivo. Profibrotic genes expression levels (fibronectin, collagen Iα, NF-KB, IL-1ß, TNF-α, SMAD4, and α-smooth muscle actin) were determined through qPCR, Western blotting, immunofluorescence staining and/or histochemical analysis in vitro and in vivo. SMAD4 targeting siRNA was further used to treat the fibroblasts in combination with miR-146a intervention to confirm its role in underlying mechanisms. RESULTS: Upregulation of miR-146a reduced the proliferation rate and profibrotic changes of rat Tenon's capsule fibroblasts induced by TGF-ß1 in vitro, and mitigated subconjunctival fibrosis to extend filtering blebs survival after GFS in vivo, where miR-146a decreased expression levels of NF-KB-SMAD4-related genes, such as fibronectin, collagen Iα, NF-KB, IL-1ß, TNF-α, SMAD4, and α-smooth muscle actin(α-SMA). Additionally, SMAD4 is a key target gene in the process of miR-146a inhibiting fibrosis. CONCLUSIONS: MiR-146a effectively reduced TGF-ß1-induced fibrosis in rat Tenon's capsule fibroblasts in vitro and in vivo, potentially through the NF-KB-SMAD4 signaling pathway. MiR-146a shows promise as a novel therapeutic target for preventing fibrosis and improving the success rate of GFS.


Sujet(s)
Fibroblastes , Fibrose , Chirurgie filtrante , Glaucome , microARN , Rat Sprague-Dawley , Animaux , microARN/métabolisme , microARN/génétique , Glaucome/anatomopathologie , Glaucome/génétique , Chirurgie filtrante/effets indésirables , Fibroblastes/métabolisme , Mâle , Capsule de Tenon/métabolisme , Capsule de Tenon/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta-1/métabolisme , Rats , Protéine Smad-4/métabolisme , Protéine Smad-4/génétique , Facteur de transcription NF-kappa B/métabolisme , Mitomycine/pharmacologie , Mitomycine/usage thérapeutique , Régulation de l'expression des gènes
18.
Cells ; 13(9)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38727311

RÉSUMÉ

Glaucoma is a heterogeneous group of optic neuropathies characterized by a progressive degeneration of the retinal ganglion cells (RGCs), leading to irreversible vision loss. Nowadays, the traditional therapeutic approach to glaucoma consists of lowering the intraocular pressure (IOP), which does not address the neurodegenerative features of the disease. Besides animal models of glaucoma, there is a considerable need for in vitro experimental models to propose new therapeutic strategies for this ocular disease. In this study, we elucidated the pathological mechanisms leading to neuroretinal R28 cell death after exposure to glutamate and hydrogen peroxide (H2O2) in order to develop new therapeutic approaches for oxidative stress-induced retinal diseases, including glaucoma. We were able to show that glutamate and H2O2 can induce a decrease in R28 cell viability in a concentration-dependent manner. A cell viability of about 42% was found after exposure to 3 mM of glutamate and about 56% after exposure to 100 µM of H2O2 (n = 4). Label-free quantitative mass spectrometry analysis revealed differential alterations of 193 and 311 proteins in R28 cells exposed to 3 mM of glutamate and 100 µM of H2O2, respectively (FDR < 1%; p < 0.05). Bioinformatics analysis indicated that the protein changes were associated with the dysregulation of signaling pathways, which was similar to those observed in glaucoma. Thus, the proteomic alteration induced by glutamate was associated with the inhibition of the PI3K/AKT signaling pathway. On the other hand, H2O2-induced toxicity in R28 cells was linked to the activation of apoptosis signaling and the inhibition of the mTOR and ERK/MAPK signaling pathways. Furthermore, the data show a similarity in the inhibition of the EIF2 and AMPK signaling pathways and the activation of the sumoylation and WNT/ß-catenin signaling pathways in both groups. Our findings suggest that the exposure of R28 cells to glutamate and H2O2 could induce glaucoma-like neurodegenerative features and potentially provide a suitable tool for the development of new therapeutic strategies for retinal diseases.


Sujet(s)
Glaucome , Acide glutamique , Peroxyde d'hydrogène , Stress oxydatif , Glaucome/métabolisme , Glaucome/anatomopathologie , Glaucome/traitement médicamenteux , Stress oxydatif/effets des médicaments et des substances chimiques , Animaux , Peroxyde d'hydrogène/pharmacologie , Acide glutamique/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Rats , Lignée cellulaire , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Cellules ganglionnaires rétiniennes/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Modèles biologiques , Humains
19.
Sci Rep ; 14(1): 10096, 2024 05 02.
Article de Anglais | MEDLINE | ID: mdl-38698014

RÉSUMÉ

Pou6f2 is a genetic connection between central corneal thickness (CCT) in the mouse and a risk factor for developing primary open-angle glaucoma. POU6F2 is also a risk factor for several conditions in humans, including glaucoma, myopia, and dyslexia. Recent findings demonstrate that POU6F2-positive retinal ganglion cells (RGCs) comprise a number of RGC subtypes in the mouse, some of which also co-stain for Cdh6 and Hoxd10. These POU6F2-positive RGCs appear to be novel of ON-OFF directionally selective ganglion cells (ooDSGCs) that do not co-stain with CART or SATB2 (typical ooDSGCs markers). These POU6F2-positive cells are sensitive to damage caused by elevated intraocular pressure. In the DBA/2J mouse glaucoma model, heavily-labeled POU6F2 RGCs decrease by 73% at 8 months of age compared to only 22% loss of total RGCs (labeled with RBPMS). Additionally, Pou6f2-/- mice suffer a significant loss of acuity and spatial contrast sensitivity along with an 11.4% loss of total RGCs. In the rhesus macaque retina, POU6F2 labels the large parasol ganglion cells that form the magnocellular (M) pathway. The association of POU6F2 with the M-pathway may reveal in part its role in human glaucoma, myopia, and dyslexia.


Sujet(s)
Dyslexie , Glaucome , Myopie , Cellules ganglionnaires rétiniennes , Animaux , Humains , Souris , Modèles animaux de maladie humaine , Dyslexie/génétique , Dyslexie/métabolisme , Dyslexie/anatomopathologie , Glaucome/anatomopathologie , Glaucome/métabolisme , Glaucome/génétique , Pression intraoculaire , Souris de lignée DBA , Souris knockout , Myopie/anatomopathologie , Myopie/métabolisme , Myopie/génétique , Cellules ganglionnaires rétiniennes/anatomopathologie , Cellules ganglionnaires rétiniennes/métabolisme , Facteurs de risque
20.
Exp Eye Res ; 244: 109927, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38750784

RÉSUMÉ

Subconjunctival fibrosis is the major cause of failure in both conventional and modern minimally invasive glaucoma surgeries (MIGSs) with subconjunctival filtration. The search for safe and effective anti-fibrotic agents is critical for improving long-term surgical outcomes. In this study, we investigated the effect of inhibiting the rapamycin-insensitive mTORC1/4E-BP1 axis on the transforming growth factor-beta 1(TGF-ß1)-induced fibrotic responses in human Tenon's fibroblasts (HTFs), as well as in a rat model of glaucoma filtration surgery (GFS). Primary cultured HTFs were treated with 3 ng/mL TGF-ß1 for 24 h, followed by treatment with 10 µM CZ415 for additional 24 h. Rapamycin (10 µM) was utilized as a control for mTORC1/4E-BP1 signaling insensitivity. The expression levels of fibrosis-associated molecules were measured using quantitative real-time PCR, Western blotting, and immunofluorescence analysis. Cell migration was assessed through the scratch wound assay. Additionally, a rat model of GFS was employed to evaluate the anti-fibrotic effect of CZ415 in vivo. Our findings indicated that both rapamycin and CZ415 treatment significantly reduced the TGF-ß1-induced cell proliferation, migration, and the expression of pro-fibrotic factors in HTFs. CZ415 also more effectively inhibited TGF-ß1-mediated collagen synthesis in HTFs compared to rapamycin. Activation of mTORC1/4E-BP signaling following TGF-ß1 exposure was highly suppressed by CZ415 but was only modestly inhibited by rapamycin. Furthermore, CZ415 was found to decrease subconjunctival collagen deposition in rats post GFS. Our results suggest that rapamycin-insensitive mTORC1/4E-BP1 signaling plays a critical role in TGF-ß1-driven collagen synthesis in HTFs. This study demonstrated that inhibition of the mTORC1/4E-BP1 axis offers superior anti-fibrotic efficacy compared to rapamycin and represents a promising target for improving the success rate of both traditional and modern GFSs.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Fibroblastes , Fibrose , Complexe-1 cible mécanistique de la rapamycine , Sirolimus , Capsule de Tenon , Facteur de croissance transformant bêta-1 , Animaux , Facteur de croissance transformant bêta-1/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/antagonistes et inhibiteurs , Humains , Rats , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Sirolimus/pharmacologie , Fibrose/métabolisme , Capsule de Tenon/métabolisme , Capsule de Tenon/effets des médicaments et des substances chimiques , Cellules cultivées , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Technique de Western , Rat Sprague-Dawley , Protéines du cycle cellulaire/métabolisme , Transduction du signal , Réaction de polymérisation en chaine en temps réel , Mâle , Glaucome/métabolisme , Glaucome/traitement médicamenteux , Glaucome/anatomopathologie , Immunosuppresseurs/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...