Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.015
Filter
1.
Acta Neuropathol Commun ; 12(1): 89, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38845058

ABSTRACT

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.


Subject(s)
Genetic Therapy , Glaucoma , tau Proteins , tau Proteins/metabolism , Animals , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/genetics , Genetic Therapy/methods , Proto-Oncogene Proteins c-akt/metabolism , Dependovirus/genetics , Disease Models, Animal , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/genetics , Retina/metabolism , Retina/pathology , MAP Kinase Signaling System/physiology , Signal Transduction/physiology , Mice , Mice, Inbred C57BL , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Phenotype
2.
Biomolecules ; 14(5)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38785932

ABSTRACT

Augmenting the natural melanocortin pathway in mouse eyes with uveitis or diabetes protects the retinas from degeneration. The retinal cells are protected from oxidative and apoptotic signals of death. Therefore, we investigated the effects of a therapeutic application of the melanocortin alpha-melanocyte-stimulating hormone (α-MSH) on an ischemia and reperfusion (I/R) model of retinal degenerative disease. Eyes were subjected to an I/R procedure and were treated with α-MSH. Retinal sections were histopathologically scored. Also, the retinal sections were immunostained for viable ganglion cells, activated Muller cells, microglial cells, and apoptosis. The I/R caused retinal deformation and ganglion cell loss that was significantly reduced in I/R eyes treated with α-MSH. While α-MSH treatment marginally reduced the number of GFAP-positive Muller cells, it significantly suppressed the density of Iba1-positive microglial cells in the I/R retinas. Within one hour after I/R, there was apoptosis in the ganglion cell layer, and by 48 h, there was apoptosis in all layers of the neuroretina. The α-MSH treatment significantly reduced and delayed the onset of apoptosis in the retinas of I/R eyes. The results demonstrate that therapeutically augmenting the melanocortin pathways preserves retinal structure and cell survival in eyes with progressive neuroretinal degenerative disease.


Subject(s)
Apoptosis , Homeostasis , Reperfusion Injury , Retina , Retinal Ganglion Cells , alpha-MSH , Animals , alpha-MSH/pharmacology , alpha-MSH/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Mice , Apoptosis/drug effects , Retina/metabolism , Retina/drug effects , Retina/pathology , Homeostasis/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Mice, Inbred C57BL , Microglia/metabolism , Microglia/drug effects , Male , Ependymoglial Cells/metabolism , Ependymoglial Cells/drug effects , Ependymoglial Cells/pathology , Disease Models, Animal , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/drug therapy
3.
Invest Ophthalmol Vis Sci ; 65(5): 15, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38717426

ABSTRACT

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.


Subject(s)
Collagen Type IV , Glaucoma , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction , Transforming Growth Factor beta , Animals , Mice , Anterior Eye Segment/metabolism , Anterior Eye Segment/pathology , Collagen Type IV/metabolism , Collagen Type IV/genetics , Disease Models, Animal , Glaucoma/metabolism , Glaucoma/genetics , Glaucoma/pathology , Intraocular Pressure/physiology , Mice, Inbred C57BL , Mutation , Optic Nerve/pathology , Optic Nerve/metabolism , Optic Nerve Diseases/metabolism , Optic Nerve Diseases/genetics , Phenotype , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Signal Transduction/physiology , Slit Lamp Microscopy , Tomography, Optical Coherence , Tonometry, Ocular , Transforming Growth Factor beta/metabolism
4.
Cell ; 187(11): 2767-2784.e23, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38733989

ABSTRACT

The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.


Subject(s)
Cerebellum , Neurons , Retina , Animals , Female , Male , Mice , Cerebellum/metabolism , Cerebellum/blood supply , Cerebellum/cytology , Ion Channels/metabolism , Mice, Inbred C57BL , Neurons/metabolism , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/metabolism , Retinal Vessels/metabolism
5.
Cell Rep Med ; 5(5): 101554, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38729157

ABSTRACT

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.


Subject(s)
Axons , Disease Models, Animal , Glaucoma , LIM-Homeodomain Proteins , Nerve Regeneration , Retinal Ganglion Cells , Transcription Factors , Animals , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Glaucoma/genetics , Glaucoma/pathology , Glaucoma/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Axons/metabolism , Axons/pathology , Mice , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Mice, Inbred C57BL , Cell Survival/genetics , Semaphorins/metabolism , Semaphorins/genetics , N-Methylaspartate/metabolism
6.
Brain Struct Funct ; 229(5): 1279-1298, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703218

ABSTRACT

ß-synuclein, a member of the synuclein family, is frequently co-expressed with α-synuclein in the neural system, where it serves to inhibit abnormal aggregation of α-synuclein in neurodegenerative diseases. Beyond its role in pathological conditions, ß-synuclein plays various functions independently of α-synuclein. In our investigation, we discovered a broader expression of ß-synuclein in the mouse retina compared to α-synuclein. This widespread pattern implies its potential significance in the retina. Through detailed examination via light- and electron-microscopic immunocytochemistry, we identified ß-synuclein expression from the inner segment (IS) and outer segment (OS) of photoreceptor cells to the ganglion cell layer (GCL). Our findings unveiled unique features, including ß-synuclein immunoreactive IS and OS of cones, higher expression in cone pedicles than in rod spherules, absence in horizontal cells, limited expression in cone bipolar dendrites and somas, higher expression in cone bipolar terminals, presence in most amacrine cells, and expression in almost majority of somas in GCL with an absence in intrinsically photosensitive retinal ganglion cell (ipRGCs) processes. Notably, all cholinergic amacrine cells express high ß- but not α-synuclein, while dopaminergic amacrine cells express α-synuclein exclusively. These distinctive expression patterns offer valuable insights for further exploration into the functions of ß-synuclein and its potential role in synuclein pathology within the retina.


Subject(s)
Mice, Inbred C57BL , Retina , Retinal Ganglion Cells , alpha-Synuclein , beta-Synuclein , Animals , Male , Mice , alpha-Synuclein/metabolism , Amacrine Cells/metabolism , beta-Synuclein/metabolism , Retina/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism
7.
Biomed Pharmacother ; 175: 116711, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735082

ABSTRACT

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.


Subject(s)
Acetylcysteine , NADP , Ocular Hypertension , Rats, Sprague-Dawley , Retinal Ganglion Cells , p38 Mitogen-Activated Protein Kinases , Animals , NADP/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Ocular Hypertension/metabolism , Ocular Hypertension/drug therapy , Ocular Hypertension/pathology , Acetylcysteine/pharmacology , Rats , Male , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/drug therapy , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Humans , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Disease Models, Animal , MAP Kinase Signaling System/drug effects , Apoptosis/drug effects , Chronic Disease , Neuroprotective Agents/pharmacology , Cells, Cultured , Lipid Peroxidation/drug effects
8.
Sci Rep ; 14(1): 10096, 2024 05 02.
Article in English | MEDLINE | ID: mdl-38698014

ABSTRACT

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.


Subject(s)
Dyslexia , Glaucoma , Myopia , Retinal Ganglion Cells , Animals , Humans , Mice , Disease Models, Animal , Dyslexia/genetics , Dyslexia/metabolism , Dyslexia/pathology , Glaucoma/pathology , Glaucoma/metabolism , Glaucoma/genetics , Intraocular Pressure , Mice, Inbred DBA , Mice, Knockout , Myopia/pathology , Myopia/metabolism , Myopia/genetics , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Risk Factors
9.
Cells ; 13(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38727311

ABSTRACT

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.


Subject(s)
Glaucoma , Glutamic Acid , Hydrogen Peroxide , Oxidative Stress , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/drug therapy , Oxidative Stress/drug effects , Animals , Hydrogen Peroxide/pharmacology , Glutamic Acid/metabolism , Cell Survival/drug effects , Rats , Cell Line , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Signal Transduction/drug effects , Models, Biological , Humans
10.
Exp Eye Res ; 244: 109931, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763353

ABSTRACT

Gene therapy is one of the strategies that may reduce or reverse progressive neurodegeneration in retinal neurodegenerative diseases. However, efficiently delivering transgenes to retinal ganglion cells (RGCs) remains hard to achieve. In this study, we innovatively investigated transduction efficiency of adeno-associated virus (AAV)-PHP.eB in murine RGCs by retro-orbital venous sinus injection. Five doses of AAV-PHP.eB-EGFP were retro-orbitally injected in venous sinus in adult C57/BL6J mice. Two weeks after administration, RGCs transduction efficiency was quantified by retinal flat-mounts and frozen section co-labeling with RGCs marker Rbpms. In addition, safety of this method was evaluated by RGCs survival rate and retinal morphology. To conform efficacy of this new method, AAV-PHP.eB-CNTF was administrated into mature mice through single retro-orbital venous injection after optic nerve crush injury to evaluate axonal elongation. Results indicated that AAV- PHP.eB readily crossed the blood-retina barrier and was able to transduce more than 90% of RGCs when total dose of virus reached 5 × 1010 vector genomes (vg). Moreover, this technique did not affect RGCs survival rate and retinal morphology. Furthermore, retro-orbital venous delivery of AAV-PHP.eB-CNTF effectively transduced RGCs, robustly promoted axonal regeneration after optic nerve crush injury. Thus, novel AAV-PHP.eB retro-orbital injection provides a minimally invasive and efficient route for transgene delivery in treatment of retinal neurodegenerative diseases.


Subject(s)
Dependovirus , Genetic Therapy , Genetic Vectors , Mice, Inbred C57BL , Retinal Ganglion Cells , Transduction, Genetic , Animals , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Mice , Dependovirus/genetics , Genetic Therapy/methods , Optic Nerve Injuries/therapy , Optic Nerve Injuries/metabolism , Disease Models, Animal , Cell Survival , Orbit/blood supply
11.
J Transl Med ; 22(1): 447, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741132

ABSTRACT

BACKGROUND: Retinal ischemia/reperfusion (RIR) is implicated in various forms of optic neuropathies, yet effective treatments are lacking. RIR leads to the death of retinal ganglion cells (RGCs) and subsequent vision loss, posing detrimental effects on both physical and mental health. Apigenin (API), derived from a wide range of sources, has been reported to exert protective effects against ischemia/reperfusion injuries in various organs, such as the brain, kidney, myocardium, and liver. In this study, we investigated the protective effect of API and its underlying mechanisms on RGC degeneration induced by retinal ischemia/reperfusion (RIR). METHODS: An in vivo model was induced by anterior chamber perfusion following intravitreal injection of API one day prior to the procedure. Meanwhile, an in vitro model was established through 1% oxygen and glucose deprivation. The neuroprotective effects of API were evaluated using H&E staining, spectral-domain optical coherence tomography (SD-OCT), Fluoro-Gold retrograde labeling, and Photopic negative response (PhNR). Furthermore, transmission electron microscopy (TEM) was employed to observe mitochondrial crista morphology and integrity. To elucidate the underlying mechanisms of API, the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, flow cytometry assay, western blot, cell counting kit-8 (CCK-8) assay, lactate dehydrogenase (LDH) assay, JC-1 kit assay, dichlorofluorescein-diacetate (DCFH-DA) assay, as well as TMRE and Mito-tracker staining were conducted. RESULTS: API treatment protected retinal inner plexiform layer (IPL) and ganglion cell complex (GCC), and improved the function of retinal ganglion cells (RGCs). Additionally, API reduced RGC apoptosis and decreased lactate dehydrogenase (LDH) release by upregulating Bcl-2 and Bcl-xL expression, while downregulating Bax and cleaved caspase-3 expression. Furthermore, API increased mitochondrial membrane potential (MMP) and decreased extracellular reactive oxygen species (ROS) production. These effects were achieved by enhancing mitochondrial function, restoring mitochondrial cristae morphology and integrity, and regulating the expression of OPA1, MFN2, and DRP1, thereby regulating mitochondrial dynamics involving fusion and fission. CONCLUSION: API protects RGCs against RIR injury by modulating mitochondrial dynamics, promoting mitochondrial fusion and fission.


Subject(s)
Apigenin , Mitochondrial Dynamics , Neuroprotective Agents , Reperfusion Injury , Retinal Ganglion Cells , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Apigenin/pharmacology , Apigenin/therapeutic use , Animals , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Mitochondrial Dynamics/drug effects , Male , Apoptosis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Models, Biological , Mice, Inbred C57BL
12.
Pol J Pathol ; 75(1): 40-53, 2024.
Article in English | MEDLINE | ID: mdl-38741428

ABSTRACT

C1q/TNF-related protein-9 (CTRP9) has been reported to play roles in several types of retinal diseases. However, the role and the potential mechanism of CTRP9 in glaucoma are still incompletely understood. The expression of CTRP9 in OGD/R-induced retinal ganglion cells (RGCs) was detected by quantitative real-time polymerase chain reaction and western blot assay. Cell proliferation was identified by cell counting Kit-8 assay. Flow cytometry, enzyme-linked immunosorbent assay and western blot assay were performed to assess cell apoptosis. Unfolded protein response (UPR), endoplasmic reticulum (ER) stress and the AMPK pathway were evaluated by western blot assay. The data showed that the expression of CTRP9 was significantly downregulated in OGD/R-induced 661W cells. OGD/R treatment reduced cell viability, promoted cell apoptosis and activated the UPR and ER stress. The overexpression of CTRP9 reversed the effects of OGD/R on 661W cell viability, apoptosis, the UPR and ER stress, as well as the AMPK pathway. However, Compound C, an inhibitor of AMPK signaling, reversed the protection of CTRP9 overexpression against injury from OGD/R in 661W cells. In summary, the results revealed that CTRP9 abated the apoptosis and UPR of OGD/R-induced RGCs by regulating the AMPK pathway, which may provide a promising target for the treatment of glaucoma.


Subject(s)
AMP-Activated Protein Kinases , Apoptosis , Endoplasmic Reticulum Stress , Retinal Ganglion Cells , Signal Transduction , Unfolded Protein Response , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Animals , AMP-Activated Protein Kinases/metabolism , Mice , Cell Line , Adiponectin/metabolism , Cell Survival , Glucose/metabolism , Glaucoma/metabolism , Glaucoma/pathology , Glycoproteins
13.
Acta Neuropathol Commun ; 12(1): 79, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773545

ABSTRACT

Neurodegenerative diseases have common underlying pathological mechanisms including progressive neuronal dysfunction, axonal and dendritic retraction, and mitochondrial dysfunction resulting in neuronal death. The retina is often affected in common neurodegenerative diseases such as Parkinson's and Alzheimer's disease. Studies have demonstrated that the retina in patients with Parkinson's disease undergoes changes that parallel the dysfunction in the brain. These changes classically include decreased levels of dopamine, accumulation of alpha-synuclein in the brain and retina, and death of dopaminergic nigral neurons and retinal amacrine cells leading to gross neuronal loss. Exploring this disease's retinal phenotype and vision-related symptoms is an important window for elucidating its pathophysiology and progression, and identifying novel ways to diagnose and treat Parkinson's disease. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is commonly used to model Parkinson's disease in animal models. MPTP is a neurotoxin converted to its toxic form by astrocytes, transported to neurons through the dopamine transporter, where it causes mitochondrial Complex I inhibition and neuron degeneration. Systemic administration of MPTP induces retinal changes in different animal models. In this study, we assessed the effects of MPTP on the retina directly via intravitreal injection in mice (5 mg/mL and 50 mg/mL to 7, 14 and 21 days post-injection). MPTP treatment induced the reduction of retinal ganglion cells-a sensitive neuron in the retina-at all time points investigated. This occurred without a concomitant loss of dopaminergic amacrine cells or neuroinflammation at any of the time points or concentrations tested. The observed neurodegeneration which initially affected retinal ganglion cells indicated that this method of MPTP administration could yield a fast and straightforward model of retinal ganglion cell neurodegeneration. To assess whether this model could be amenable to neuroprotection, mice were treated orally with nicotinamide (a nicotinamide adenine dinucleotide precursor) which has been demonstrated to be neuroprotective in several retinal ganglion cell injury models. Nicotinamide was strongly protective following intravitreal MPTP administration, further supporting intravitreal MPTP use as a model of retinal ganglion cell injury. As such, this model could be utilized for testing neuroprotective treatments in the context of Parkinson's disease and retinal ganglion cell injury.


Subject(s)
Mice, Inbred C57BL , Neuroprotective Agents , Niacinamide , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Niacinamide/pharmacology , Niacinamide/administration & dosage , Neuroprotective Agents/pharmacology , Neuroprotective Agents/administration & dosage , Male , Mice , Administration, Oral , Intravitreal Injections , Disease Models, Animal , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/drug therapy , MPTP Poisoning/pathology , MPTP Poisoning/metabolism , MPTP Poisoning/drug therapy
14.
Invest Ophthalmol Vis Sci ; 65(5): 36, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38776115

ABSTRACT

Purpose: The purpose of this study was to investigate the protective effect of CD38 deletion on retinal ganglion cells (RGCs) in a mouse retinal ischemia/reperfusion (I/R) model and an optic nerve crush (ONC) model, and to elucidate the underlying molecular mechanisms. Methods: Retinal I/R and ONC models were constructed in mice. PCR was used to identify the deletion of CD38 gene in mice, hematoxylin and eosin (H&E) staining was used to evaluate the changes in retinal morphology, and electroretinogram (ERG) was used to evaluate the changes in retinal function. The survival of RGCs and activation of retinal macroglia were evaluated by immunofluorescence staining. The expression of Sirt1, CD38, Ac-p65, Ac-p53, TNF-α, IL-1ß, and Caspase3 proteins in the retina was further evaluated by protein imprinting. Results: In retinal I/R and ONC models, CD38 deficiency reduced the loss of RGCs and activation of macroglia and protected the retinal function. CD38 deficiency increased the concentration of NAD+, reduced the degree of acetylation of NF-κB p65 and p53, and reduced expression of the downstream inflammatory cytokines TNFα, IL-1ß, and apoptotic protein Caspase3 in the retina in the ONC model. Intraperitoneal injection of the Sirt1 inhibitor EX-527 partially counteracted the effects of CD38 deficiency, suggesting that CD38 deficiency acts at least in part through the NAD+/Sirt1 pathway. Conclusions: CD38 plays an important role in the pathogenesis of retinal I/R and ONC injury. CD38 deletion protects RGCs by attenuating inflammatory responses and apoptosis through the NAD+/Sirt1 pathway.


Subject(s)
ADP-ribosyl Cyclase 1 , Disease Models, Animal , Mice, Inbred C57BL , NAD , Optic Nerve Injuries , Reperfusion Injury , Retinal Ganglion Cells , Sirtuin 1 , Animals , Sirtuin 1/metabolism , Sirtuin 1/genetics , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , ADP-ribosyl Cyclase 1/metabolism , ADP-ribosyl Cyclase 1/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Mice , NAD/metabolism , Optic Nerve Injuries/metabolism , Electroretinography , Nerve Crush , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Male , Signal Transduction/physiology
15.
Exp Neurol ; 377: 114810, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38714284

ABSTRACT

Most projection neurons, including retinal ganglion cells (RGCs), undergo cell death after axotomy proximal to the cell body. Specific RGC subtypes, such as ON-OFF direction selective RGCs (ooDSGCs) are particularly vulnerable, whereas intrinsically photosensitive RGCs (ipRGCs) exhibit resilience to axonal injury. Through the application of RNA sequencing and fluorescent in situ hybridization, we show that the expression of chloride intracellular channel protein 1 and 4 (Clic1 and Clic4) are highly increased in the ooDSGCs after axonal injury. Toward determining a gene's role in RGCs, we optimized the utility and efficacy of adenovirus associated virus (AAV)-retro expressing short hairpin RNA (shRNA). Injection of AAV2-retro into the superior colliculus results in efficient shRNA expression in RGCs. Incorporating histone H2B gene fused with mGreenLantern results in bright nuclear reporter expression, thereby enhancing single RGC identification and cell quantitation in live retinas. Lastly, we demonstrate that AAV2-retro mediated knockdown of both Clic1 and Clic4 promotes RGC survival after injury. Our findings establish an integrated use of AAV2-retro-shRNA and real-time fundus imaging and reveal CLICs' contribution to RGC death.


Subject(s)
Cell Death , Chloride Channels , Dependovirus , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/metabolism , Dependovirus/genetics , Chloride Channels/genetics , Chloride Channels/metabolism , Cell Death/physiology , Mice , Mice, Inbred C57BL , Male , RNA, Small Interfering/genetics
16.
Mol Ther ; 32(6): 1760-1778, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38659223

ABSTRACT

Glaucoma is characterized by the progressive degeneration of retinal ganglion cells (RGCs) and their axons, and its risk increases with aging. Yet comprehensive insights into the complex mechanisms are largely unknown. Here, we found that anti-aging molecule Sirt6 was highly expressed in RGCs. Deleting Sirt6 globally or specifically in RGCs led to progressive RGC loss and optic nerve degeneration during aging, despite normal intraocular pressure (IOP), resembling a phenotype of normal-tension glaucoma. These detrimental effects were potentially mediated by accelerated RGC senescence through Caveolin-1 upregulation and by the induction of mitochondrial dysfunction. In mouse models of high-tension glaucoma, Sirt6 level was decreased after IOP elevation. Genetic overexpression of Sirt6 globally or specifically in RGCs significantly attenuated high tension-induced degeneration of RGCs and their axons, whereas partial or RGC-specific Sirt6 deletion accelerated RGC loss. Importantly, therapeutically targeting Sirt6 with pharmacological activator or AAV2-mediated gene delivery ameliorated high IOP-induced RGC degeneration. Together, our studies reveal a critical role of Sirt6 in preventing RGC and optic nerve degeneration during aging and glaucoma, setting the stage for further exploration of Sirt6 activation as a potential therapy for glaucoma.


Subject(s)
Aging , Disease Models, Animal , Glaucoma , Optic Nerve , Retinal Ganglion Cells , Sirtuins , Animals , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Mice , Sirtuins/metabolism , Sirtuins/genetics , Glaucoma/metabolism , Glaucoma/genetics , Glaucoma/pathology , Glaucoma/etiology , Optic Nerve/metabolism , Optic Nerve/pathology , Aging/metabolism , Aging/genetics , Intraocular Pressure , Humans , Axons/metabolism , Axons/pathology , Mice, Knockout , Nerve Degeneration/metabolism
17.
Exp Eye Res ; 243: 109907, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38649019

ABSTRACT

Sleep loss is common in modern society and is increasingly associated with eye diseases. However, the precise effects of sleep loss on retinal structure and function, particularly on the retinal circadian system, remain largely unexplored. This study investigates these effects using a chronic sleep deprivation (CSD) model in mice. Our investigation reveals that CSD significantly alters the retinal circadian transcriptome, leading to remarkable changes in the temporal patterns of enriched pathways. This perturbation extends to metabolic and immune-related transcriptomes, coupled with an accumulation of reactive oxygen species in the retina. Notably, CSD rhythmically affects the thickness of the ganglion cell complex, along with diurnal shifts in microglial migration and morphology within the retina. Most critically, we observe a marked decrease in both scotopic and photopic retinal function under CSD conditions. These findings underscore the broad impact of sleep deprivation on retinal health, highlighting its role in altering circadian gene expression, metabolism, immune response, and structural integrity. Our study provides new insights into the broader impact of sleep loss on retinal health.


Subject(s)
Circadian Rhythm , Mice, Inbred C57BL , Retina , Sleep Deprivation , Transcriptome , Animals , Sleep Deprivation/physiopathology , Sleep Deprivation/metabolism , Sleep Deprivation/genetics , Mice , Circadian Rhythm/physiology , Male , Retina/metabolism , Retina/physiopathology , Disease Models, Animal , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Electroretinography , Gene Expression Regulation , Chronic Disease
18.
J Ocul Pharmacol Ther ; 40(5): 309-324, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38603587

ABSTRACT

Purpose: Glaucoma is a complex degenerative optic neuropathy characterized by loss of retinal ganglion cells (RGCs) leading to irreversible vision loss and blindness. Solanum nigrum has been used for decades in traditional medicine system. However, no extensive studies were reported on its antiglaucoma properties. Therefore, this study was designed to investigate the neuroprotective effects of S. nigrum extract on RGC against glaucoma rat model. Methods: High performance liquid chromatography and liquid chromatography tandem mass spectrometry was used to analyze the phytochemical profile of aqueous extract of S. nigrum (AESN). In vitro, {3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide} (MTT) and H2DCFDA assays were used to determine cell viability and reactive oxygen species (ROS) production in Statens Seruminstitut Rabbit Cornea cells. In vivo, AESN was orally administered to carbomer-induced rats for 4 weeks. Intraocular pressure, antioxidant levels, and electrolytes were determined. Histopathological and immunohistochemical analysis was carried out to evaluate the neurodegeneration of RGC. Results: MTT assay showed AESN exhibited greater cell viability and minimal ROS production at 10 µg/mL. Slit lamp and funduscopy confirmed glaucomatous changes in carbomer-induced rats. Administration of AESN showed minimal peripheral corneal vascularization and restored histopathological alterations such as minimal loss of corneal epithelium and moderate narrowing of the iridocorneal angle. Immunohistochemistry analysis showed increased expression of positive BRN3A cells and decreased matrix metalloproteinase (MMP)-9 activation in retina and cornea, whereas western blot analysis revealed downregulation of extracellular matrix proteins (COL-1 and MMP-9) in AESN-treated rats compared with the diseased group rats. Conclusions: AESN protects RGC loss through remodeling of MMPs and, therefore, can be used for the development of novel neurotherapeutics for the treatment of glaucoma.


Subject(s)
Cell Survival , Disease Models, Animal , Extracellular Matrix , Glaucoma , Neuroprotective Agents , Plant Extracts , Reactive Oxygen Species , Retinal Ganglion Cells , Solanum nigrum , Animals , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Glaucoma/drug therapy , Glaucoma/pathology , Glaucoma/metabolism , Rats , Solanum nigrum/chemistry , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Cell Survival/drug effects , Male , Rabbits , Intraocular Pressure/drug effects , Cell Death/drug effects , Rats, Sprague-Dawley
19.
PLoS Genet ; 20(4): e1011139, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38669217

ABSTRACT

As essential components of gene expression networks, transcription factors regulate neural circuit assembly. The homeobox transcription factor encoding gene, gs homeobox 1 (gsx1), is expressed in the developing visual system; however, no studies have examined its role in visual system formation. In zebrafish, retinal ganglion cell (RGC) axons that transmit visual information to the brain terminate in ten arborization fields (AFs) in the optic tectum (TeO), pretectum (Pr), and thalamus. Pretectal AFs (AF1-AF9) mediate distinct visual behaviors, yet we understand less about their development compared to AF10 in the TeO. Using gsx1 zebrafish mutants, immunohistochemistry, and transgenic lines, we observed that gsx1 is required for vesicular glutamate transporter, Tg(slc17a6b:DsRed), expression in the Pr, but not overall neuron number. gsx1 mutants have normal eye morphology, yet they exhibit impaired visual ability during prey capture. RGC axon volume in the gsx1 mutant Pr and TeO is reduced, and AF7 that is active during feeding is missing which is consistent with reduced hunting performance. Timed laser ablation of Tg(slc17a6b:DsRed)-positive cells reveals that they are necessary for AF7 formation. This work is the first to implicate gsx1 in establishing cell identity and functional neural circuits in the visual system.


Subject(s)
Animals, Genetically Modified , Gene Expression Regulation, Developmental , Homeodomain Proteins , Retinal Ganglion Cells , Zebrafish Proteins , Zebrafish , Animals , Axons/metabolism , Axons/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mutation , Retinal Ganglion Cells/metabolism , Superior Colliculi/metabolism , Superior Colliculi/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism , Visual Pathways/growth & development , Visual Pathways/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
Acta Neuropathol Commun ; 12(1): 65, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649962

ABSTRACT

The progressive and irreversible degeneration of retinal ganglion cells (RGCs) and their axons is the major characteristic of glaucoma, a leading cause of irreversible blindness worldwide. Nicotinamide adenine dinucleotide (NAD) is a cofactor and metabolite of redox reaction critical for neuronal survival. Supplementation with nicotinamide (NAM), a precursor of NAD, can confer neuroprotective effects against glaucomatous damage caused by an age-related decline of NAD or mitochondrial dysfunction, reflecting the high metabolic activity of RGCs. However, oral supplementation of drug is relatively less efficient in terms of transmissibility to RGCs compared to direct delivery methods such as intraocular injection or delivery using subconjunctival depots. Neither method is ideal, given the risks of infection and subconjunctival scarring without novel techniques. By contrast, extracellular vesicles (EVs) have advantages as a drug delivery system with low immunogeneity and tissue interactions. We have evaluated the EV delivery of NAM as an RGC protective agent using a quantitative assessment of dendritic integrity using DiOlistics, which is confirmed to be a more sensitive measure of neuronal health in our mouse glaucoma model than the evaluation of somatic loss via the immunostaining method. NAM or NAM-loaded EVs showed a significant neuroprotective effect in the mouse retinal explant model. Furthermore, NAM-loaded EVs can penetrate the sclera once deployed in the subconjunctival space. These results confirm the feasibility of using subconjunctival injection of EVs to deliver NAM to intraocular targets.


Subject(s)
Extracellular Vesicles , Glaucoma , Mice, Inbred C57BL , Neuroprotective Agents , Niacinamide , Retinal Ganglion Cells , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/drug effects , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Niacinamide/administration & dosage , Niacinamide/pharmacology , Mice , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Glaucoma/metabolism , Glaucoma/drug therapy , Neuroprotection/drug effects , Sclera/metabolism , Sclera/drug effects , Drug Delivery Systems/methods , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...