Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 748
Filter
1.
Invest Ophthalmol Vis Sci ; 65(10): 3, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087933

ABSTRACT

Purpose: Primary open-angle glaucoma (POAG) is a leading cause of blindness, and its primary risk factor is elevated intraocular pressure (IOP) due to pathologic changes in the trabecular meshwork (TM). We previously showed that there is a cross-inhibition between TGFß and Wnt signaling pathways in the TM. In this study, we determined if activation of the Wnt signaling pathway using small-molecule Wnt activators can inhibit TGFß2-induced TM changes and ocular hypertension (OHT). Methods: Primary human TM (pHTM) cells and transduced SBE-GTM3 cells were treated with or without Wnt and/or TGFß signaling activators and used for luciferase assays; for the extraction of whole-cell lysate, conditioned medium, cytosolic proteins, and nuclear proteins for Western immunoblotting (WB); or for immunofluorescent staining. Human donor eyes were perfusion cultured to study the effect of Wnt activators on IOP. Results: We found that the small-molecule Wnt activators (GSK3ß inhibitors) (BIO, SB216763, and CHIR99021) activated canonical Wnt signaling in pHTM cells without toxicity at tested concentrations. This activation inhibited TGFß signaling as well as TGFß2-induced extracellular matrix deposition and formation of cross-linked actin networks in pHTM cells or SBE-GTM3 cells. We also observed nuclear translocation of both Smad4 and ß-catenin in pHTM cells, which suggested that the cross-inhibition between the TGFß and Wnt signaling pathways may occur in the nucleus. Using our ex vivo model, we found that CHIR99021 inhibited TGFß2-induced OHT in perfusion-cultured human eyes. Conclusions: Our results showed that small-molecule Wnt activators have the potential for treating TGFß signaling-induced OHT in patients with POAG.


Subject(s)
Glaucoma, Open-Angle , Glycogen Synthase Kinase 3 beta , Intraocular Pressure , Trabecular Meshwork , Humans , Trabecular Meshwork/metabolism , Trabecular Meshwork/drug effects , Intraocular Pressure/physiology , Intraocular Pressure/drug effects , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/drug therapy , Cells, Cultured , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Blotting, Western , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , Ocular Hypertension/metabolism , Ocular Hypertension/drug therapy , Signal Transduction , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta2/pharmacology
2.
FASEB J ; 38(15): e23848, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39092889

ABSTRACT

Glucocorticoid use may cause elevated intraocular pressure, leading to the development of glucocorticoid-induced glaucoma (GIG). However, the mechanism of GIG development remains incompletely understood. In this study, we subjected primary human trabecular meshwork cells (TMCs) and mice to dexamethasone treatment to mimic glucocorticoid exposure. The myofibroblast transdifferentiation of TMCs was observed in cellular and mouse models, as well as in human trabecular mesh specimens. This was demonstrated by the cytoskeletal reorganization, alterations in cell morphology, heightened transdifferentiation markers, increased extracellular matrix deposition, and cellular dysfunction. Knockdown of Rho guanine nucleotide exchange factor 26 (ARHGEF26) expression ameliorated dexamethasone-induced changes in cell morphology and upregulation of myofibroblast markers, reversed dysfunction and extracellular matrix deposition in TMCs, and prevented the development of dexamethasone-induced intraocular hypertension. And, this process may be related to the TGF-ß pathway. In conclusion, glucocorticoids induced the myofibroblast transdifferentiation in TMCs, which played a crucial role in the pathogenesis of GIG. Inhibition of ARHGEF26 expression protected TMCs by reversing myofibroblast transdifferentiation. This study demonstrated the potential of reversing the myofibroblast transdifferentiation of TMCs as a new target for treating GIG.


Subject(s)
Cell Transdifferentiation , Dexamethasone , Glaucoma , Myofibroblasts , Rho Guanine Nucleotide Exchange Factors , Trabecular Meshwork , Dexamethasone/pharmacology , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism , Trabecular Meshwork/cytology , Cell Transdifferentiation/drug effects , Animals , Humans , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/cytology , Mice , Rho Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Glaucoma/pathology , Glaucoma/metabolism , Cells, Cultured , Glucocorticoids/pharmacology , Mice, Inbred C57BL , Male
3.
Invest Ophthalmol Vis Sci ; 65(8): 1, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949632

ABSTRACT

Purpose: Glucocorticoid-induced glaucoma (GIG) is a prevalent complication associated with glucocorticoids (GCs), resulting in irreversible blindness. GIG is characterized by the abnormal deposition of extracellular matrix (ECM) in the trabecular meshwork (TM), elevation of intraocular pressure (IOP), and loss of retinal ganglion cells (RGCs). The objective of this study is to investigate the effects of nicotinamide riboside (NR) on TM in GIG. Methods: Primary human TM cells (pHTMs) and C57BL/6J mice responsive to GCs were utilized to establish in vitro and in vivo GIG models, respectively. The study assessed the expression of ECM-related proteins in TM and the functions of pHTMs to reflect the effects of NR. Mitochondrial morphology and function were also examined in the GIG cell model. GIG progression was monitored through IOP, RGCs, and mitochondrial morphology. Intracellular nicotinamide adenine dinucleotide (NAD+) levels of pHTMs were enzymatically assayed. Results: NR significantly prevented the expression of ECM-related proteins and alleviated dysfunction in pHTMs after dexamethasone treatment. Importantly, NR protected damaged ATP synthesis, preventing overexpression of mitochondrial reactive oxygen species (ROS), and also protect against decreased mitochondrial membrane potential induced by GCs in vitro. In the GIG mouse model, NR partially prevented the elevation of IOP and the loss of RGCs. Furthermore, NR effectively suppressed the excessive expression of ECM-associated proteins and mitigated mitochondrial damage in vivo. Conclusions: Based on the results, NR effectively enhances intracellular levels of NAD+, thereby mitigating abnormal ECM deposition and TM dysfunction in GIG by attenuating mitochondrial damage induced by GCs. Thus, NR has promising potential as a therapeutic candidate for GIG treatment.


Subject(s)
Disease Models, Animal , Extracellular Matrix , Glaucoma , Glucocorticoids , Intraocular Pressure , Mice, Inbred C57BL , Mitochondria , Niacinamide , Pyridinium Compounds , Trabecular Meshwork , Animals , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Pyridinium Compounds/pharmacology , Glucocorticoids/toxicity , Mitochondria/metabolism , Mitochondria/drug effects , Mice , Glaucoma/metabolism , Glaucoma/drug therapy , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Intraocular Pressure/drug effects , Humans , Trabecular Meshwork/metabolism , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology , Cells, Cultured , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Reactive Oxygen Species/metabolism , Dexamethasone/pharmacology , Male
4.
Exp Cell Res ; 440(1): 114137, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38897410

ABSTRACT

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.


Subject(s)
Glaucoma , Mitochondria , NAD , Trabecular Meshwork , Trabecular Meshwork/metabolism , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/drug therapy , NAD/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Intraocular Pressure/drug effects , Cell Survival/drug effects , ADP-ribosyl Cyclase 1/metabolism , ADP-ribosyl Cyclase 1/genetics , Cell Line , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Dexamethasone/pharmacology , Cells, Cultured
5.
Cells ; 13(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38920689

ABSTRACT

Primary open-angle glaucoma (POAG) is a progressive optic neuropathy with a complex, multifactorial aetiology. Raised intraocular pressure (IOP) is the most important clinically modifiable risk factor for POAG. All current pharmacological agents target aqueous humour dynamics to lower IOP. Newer therapeutic agents are required as some patients with POAG show a limited therapeutic response or develop ocular and systemic side effects to topical medication. Elevated IOP in POAG results from cellular and molecular changes in the trabecular meshwork driven by increased levels of transforming growth factor ß (TGFß) in the anterior segment of the eye. Understanding how TGFß affects both the structural and functional changes in the outflow pathway and IOP is required to develop new glaucoma therapies that target the molecular pathology in the trabecular meshwork. In this study, we evaluated the effects of TGF-ß1 and -ß2 treatment on miRNA expression in cultured human primary trabecular meshwork cells. Our findings are presented in terms of specific miRNAs (miRNA-centric), but given miRNAs work in networks to control cellular pathways and processes, a pathway-centric view of miRNA action is also reported. Evaluating TGFß-responsive miRNA expression in trabecular meshwork cells will further our understanding of the important pathways and changes involved in the pathogenesis of glaucoma and could lead to the development of miRNAs as new therapeutic modalities in glaucoma.


Subject(s)
MicroRNAs , Trabecular Meshwork , Trabecular Meshwork/metabolism , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/pathology , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Cells, Cultured , Gene Expression Regulation/drug effects , Intraocular Pressure/drug effects
6.
FASEB J ; 38(10): e23651, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38752537

ABSTRACT

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.


Subject(s)
Autophagy , DEAD Box Protein 58 , Glaucoma , Intraocular Pressure , Trabecular Meshwork , Animals , Female , Humans , Male , Mice , Aortic Diseases , Autophagy/drug effects , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , Dental Enamel Hypoplasia , Glaucoma/pathology , Glaucoma/metabolism , Glaucoma/genetics , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/metabolism , Interferon-beta/metabolism , Intraocular Pressure/genetics , Metacarpus/abnormalities , Mice, Inbred C57BL , Muscular Diseases , Mutation , Odontodysplasia , Optic Atrophy/genetics , Optic Atrophy/metabolism , Optic Atrophy/pathology , Osteoporosis , Pedigree , Receptors, Immunologic , Trabecular Meshwork/metabolism , Trabecular Meshwork/drug effects , Vascular Calcification
7.
Exp Eye Res ; 244: 109939, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38789021

ABSTRACT

Transforming growth factor-ß2 (TGF-ß2) induced fibrogenic changes in human trabecular meshwork (HTM) cells have been implicated in trabecular meshwork (TM) damage and intraocular pressure (IOP) elevation in primary open-angle glaucoma (POAG) patients. Silibinin (SIL) exhibited anti-fibrotic properties in various organs and tissues. This study aimed to assess the effects of SIL on the TGF-ß2-treated HTM cells and to elucidate the underlying mechanisms. Our study found that SIL effectively inhibited HTM cell proliferation, attenuated TGF-ß2-induced cell migration, and mitigated TGF-ß2-induced reorganization of both actin and vimentin filaments. Moreover, SIL suppressed the expressions of fibronectin (FN), collagen type I alpha 1 chain (COL1A1), and alpha-smooth muscle actin (α-SMA) in the TGF-ß2-treated HTM cells. RNA sequencing indicated that SIL interfered with the phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB, also known as AKT) signaling pathway, extracellular matrix (ECM)-receptor interaction, and focal adhesion in the TGF-ß2-treated HTM cells. Western blotting demonstrated SIL inhibited the activation of Janus kinase 2 (JAK2)/signal transducers and activators of transcription 3 (STAT3) and the downstream PI3K/AKT signaling pathways induced by TGF-ß2, potentially contributing to its inhibitory effects on ECM protein production in the TGF-ß2-treated HTM cells. Our study demonstrated the ability of SIL to inhibit TGF-ß2-induced fibrogenic changes in HTM cells. SIL could be a potential IOP-lowering agent by reducing the fibrotic changes in the TM tissue of POAG patients, which warrants further investigation through additional animal and clinical studies.


Subject(s)
Cell Movement , Cell Proliferation , Signal Transduction , Silybin , Trabecular Meshwork , Humans , Antioxidants/pharmacology , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibrosis , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/drug therapy , Glaucoma, Open-Angle/pathology , Janus Kinase 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Silybin/pharmacology , Silymarin/pharmacology , STAT3 Transcription Factor/metabolism , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism , Trabecular Meshwork/pathology , Transforming Growth Factor beta2/pharmacology , Transforming Growth Factor beta2/metabolism
8.
Int Ophthalmol ; 44(1): 229, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38795168

ABSTRACT

BACKGROUND: The multifunctional profibrotic cytokine transforming growth factor-beta2 (TGF-ß2) is implicated in the pathophysiology of primary open angle glaucoma. Paeoniflorin (PAE) is a monoterpene glycoside with multiple pharmacological efficacies, such as antioxidant, anti-fibrotic, and anti-inflammatory properties. Studies have demonstrated that paeoniflorin protects human corneal epithelial cells, retinal pigment epithelial cells, and retinal microglia from damage. Here, the biological role of PAE in TGF-ß2-dependent remodeling of the extracellular matrix (ECM) within the trabecular meshwork (TM) microenvironment. METHODS: Primary or transformed (GTM3) human TM (HTM) cells conditioned in serum-free media were incubated with TGF-ß2 (5 ng/mL). PAE (300 µM) was added to serum-starved confluent cultures of HTM cells for 2 h, followed by incubation with TGF-ß2 for 22 h. SB-431542, a TGF-ß receptor inhibitor (10 µM), was used as a positive control. The levels of intracellular ROS were evaluated by CellROX green dye. Western blotting was used to measure the levels of TGF-ß2/Smad2/3 signaling-related molecules. Collagen 1α1, collagen 4α1, and connective tissue growth factor (CTGF) expression was evaluated by RT-qPCR. Immunofluorescence assay was conducted to measure collagen I/IV expression in HTM cells. Phalloidin staining assay was conducted for evaluating F-actin stress fiber formation in the cells. RESULTS: PAE attenuated TGF-ß2-induced oxidative stress and suppressed TGF-ß2-induced Smad2/3 signaling in primary or transformed HTM cells. Additionally, PAE repressed TGF-ß2-induced upregulation of collagen 1α1, collagen 4α1, and CTGF expression and reduced TGF-ß2-mediated collagen I/IV expression and of F-actin stress fiber formation in primary or transformed HTM cells. CONCLUSION: PAE alleviates TGF-ß2-induced ECM deposition and oxidative stress in HTM cells through inactivation of Smad2/3 signaling.


Subject(s)
Extracellular Matrix , Glucosides , Monoterpenes , Oxidative Stress , Trabecular Meshwork , Transforming Growth Factor beta2 , Humans , Oxidative Stress/drug effects , Monoterpenes/pharmacology , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology , Glucosides/pharmacology , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism , Trabecular Meshwork/pathology , Cells, Cultured , Signal Transduction/drug effects , Blotting, Western
9.
Am J Physiol Cell Physiol ; 326(5): C1505-C1519, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38557355

ABSTRACT

Glaucoma is a blinding disease. Reduction of intraocular pressure (IOP) is the mainstay of treatment, but current drugs show side effects or become progressively ineffective, highlighting the need for novel compounds. We have synthesized a family of perhydro-1,4-oxazepine derivatives of digoxin, the selective inhibitor of Na,K-ATPase. The cyclobutyl derivative (DcB) displays strong selectivity for the human α2 isoform and potently reduces IOP in rabbits. These observations appeared consistent with a hypothesis that in ciliary epithelium DcB inhibits the α2 isoform of Na,K-ATPase, which is expressed strongly in nonpigmented cells, reducing aqueous humor (AH) inflow. This paper extends assessment of efficacy and mechanism of action of DcB using an ocular hypertensive nonhuman primate model (OHT-NHP) (Macaca fascicularis). In OHT-NHP, DcB potently lowers IOP, in both acute (24 h) and extended (7-10 days) settings, accompanied by increased aqueous humor flow rate (AFR). By contrast, ocular normotensive animals (ONT-NHP) are poorly responsive to DcB, if at all. The mechanism of action of DcB has been analyzed using isolated porcine ciliary epithelium and perfused enucleated eyes to study AH inflow and AH outflow facility, respectively. 1) DcB significantly stimulates AH inflow although prior addition of 8-Br-cAMP, which raises AH inflow, precludes additional effects of DcB. 2) DcB significantly increases AH outflow facility via the trabecular meshwork (TM). Taken together, the data indicate that the original hypothesis on the mechanism of action must be revised. In the OHT-NHP, and presumably other species, DcB lowers IOP by increasing AH outflow facility rather than by decreasing AH inflow.NEW & NOTEWORTHY When applied topically, a cyclobutyl derivative of digoxin (DcB) potently reduces intraocular pressure in an ocular hypertensive nonhuman primate model (Macaca fascicularis), associated with increased aqueous humor (AH) flow rate (AFR). The mechanism of action of DcB involves increased AH outflow facility as detected in enucleated perfused porcine eyes and, in parallel, increased (AH) inflow as detected in isolated porcine ciliary epithelium. DcB might have potential as a drug for the treatment of open-angle human glaucoma.


Subject(s)
Aqueous Humor , Digoxin , Intraocular Pressure , Macaca fascicularis , Ocular Hypertension , Animals , Intraocular Pressure/drug effects , Digoxin/pharmacology , Aqueous Humor/metabolism , Aqueous Humor/drug effects , Ocular Hypertension/drug therapy , Ocular Hypertension/physiopathology , Ocular Hypertension/metabolism , Disease Models, Animal , Glaucoma/drug therapy , Glaucoma/metabolism , Glaucoma/physiopathology , Rabbits , Humans , Ciliary Body/drug effects , Ciliary Body/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Male , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism
10.
J Cell Physiol ; 238(3): 631-646, 2023 03.
Article in English | MEDLINE | ID: mdl-36727620

ABSTRACT

A common adverse response to the clinical use of glucocorticoids (GCs) is elevated intraocular pressure (IOP) which is a major risk factor for glaucoma. Elevated IOP arises due to impaired outflow of aqueous humor (AH) through the trabecular meshwork (TM). Although GC-induced changes in actin cytoskeletal dynamics, contractile characteristics, and cell adhesive interactions of TM cells are believed to influence AH outflow and IOP, the molecular mechanisms mediating changes in these cellular characteristics are poorly understood. Our studies focused on evaluating changes in the cytoskeletal and cytoskeletal-associated protein (cytoskeletome) profile of human TM cells treated with dexamethasone (Dex) using label-free mass spectrometric quantification, identified elevated levels of specific proteins known to regulate actin stress fiber formation, contraction, actin networks crosslinking, cell adhesion, and Wnt signaling, including LIMCH1, ArgBP2, CNN3, ITGBL1, CTGF, palladin, FAT1, DIAPH2, EPHA4, SIPA1L1, and GPC4. Several of these proteins colocalized with the actin cytoskeleton and underwent alterations in distribution profile in TM cells treated with Dex, and an inhibitor of Abl/Src kinases. Wnt/Planar Cell Polarity (PCP) signaling agonists-Wnt5a and 5b were detected prominently in the cytoskeletome fraction of TM cells, and studies using siRNA to suppress expression of glypican-4 (GPC4), a known modulator of the Wnt/PCP pathway revealed that GPC4 deficiency impairs Dex induced actin stress fiber formation, and activation of c-Jun N-terminal Kinase (JNK) and Rho kinase. Additionally, while Dex augmented, GPC4 deficiency suppressed the formation of actin stress fibers in TM cells in the presence of Dex and Wnt5a. Taken together, these results identify the GPC4-dependent Wnt/PCP signaling pathway as one of the crucial upstream regulators of Dex induced actin cytoskeletal reorganization and cell adhesion in TM cells, opening an opportunity to target the GPC4/Wnt/PCP pathway for treatment of ocular hypertension in glaucoma.


Subject(s)
Actins , Cytoskeletal Proteins , Cytoskeleton , Dexamethasone , Glucocorticoids , Glypicans , Trabecular Meshwork , Humans , Actins/metabolism , Cells, Cultured , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dexamethasone/pharmacology , Glaucoma/metabolism , Glaucoma/pathology , Glucocorticoids/pharmacology , Glypicans/deficiency , Glypicans/metabolism , Intraocular Pressure , Trabecular Meshwork/cytology , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism , Wnt Signaling Pathway/drug effects , Cytoskeleton/metabolism , Cell Polarity/drug effects , rho-Associated Kinases/metabolism , Stress Fibers/drug effects , Cell Adhesion/drug effects
11.
Exp Eye Res ; 216: 108914, 2022 03.
Article in English | MEDLINE | ID: mdl-34979099

ABSTRACT

Glaucoma, characterized by ocular hypertension, is the second most common cause of vision loss worldwide. The potential mechanism, however, has yet to be elucidated. This study aimed to assess the proteomic changes in the trabecular meshwork (TM) in an observational animal model of Dexamethasone (DEX)-induced OHT. OHT was induced in Wistar rats by applying DEX topically to both eyes for 28 days. Intraocular pressure (IOP) was evaluated and TM protein expressions and protein identification were performed by a TMT-based method for comparing the changes in proteins between DEX-induced OHT and the control group. The results showed that average IOP was elevated significantly in rats of the DEX-induced OHT group compared to controls. Further, a total of 4,804 proteins in the control and DEX-induced OHT group were determined and 4,064 proteins were quantified via TMT proteomics. In total, 292 significantly abundant proteins (173 downregulated and 119 upregulated) were identified between the two groups. Proteins associated with vision, including Crystallin related proteins, filensin, rhodopsin, recoverin, phosducin were lowered in the DEX-induced OHT group relative to the control group. In summary, DEX induced extensive changes in the protein expression of TM tissue. These proteins were found to be candidate biomarkers for personalized treatment and diagnostic research in the future for improving visual health.


Subject(s)
Biomarkers/metabolism , Dexamethasone/adverse effects , Eye Proteins/metabolism , Glucocorticoids/adverse effects , Ocular Hypertension/chemically induced , Trabecular Meshwork/drug effects , Animals , Chromatography, High Pressure Liquid , Disease Models, Animal , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Female , Intraocular Pressure/drug effects , Male , Ocular Hypertension/metabolism , Proteomics , Rats , Rats, Wistar , Tandem Mass Spectrometry , Tonometry, Ocular , Trabecular Meshwork/metabolism
12.
Exp Eye Res ; 216: 108935, 2022 03.
Article in English | MEDLINE | ID: mdl-35033558

ABSTRACT

PURPOSE: Cells in the trabecular meshwork sense and respond to a myriad of physical forces through a process known as mechanotransduction. Whilst the effect of substratum stiffness or stretch on TM cells have been investigated in the context of transforming growth factor (TGF-ß), Wnt and YAP/TAZ pathways, the role of Notch signaling, an evolutionarily conserved pathway, recently implicated in mechanotransduction, has not been investigated in trabecular meshwork (TM) cells. Here, we compare the endogenous expression of Notch pathway molecules in TM cells from glaucomatous and non-glaucomatous donors, segmental flow regions, and when subjected to cyclical strain, or grown on hydrogels of varying rigidity. METHODS: Primary TM from glaucomatous (GTM), non-glaucomatous (NTM) donors, and from segmental flow regions [high flow (HF), low flow (LF)], were utilized between passages 2-6. Cells were (i) plated on tissue culture plastic, (ii) subjected to cyclical strain (6 h and 24 h), or (iii) cultured on 3 kPa and 80 kPa hydrogels. mRNA levels of Notch receptors/ligands/effectors in the TM cells was determined by qRT-PCR. Phagocytosis was determined as a function of substratum stiffness in NTM-HF/LF cells in the presence or absence of 100 nM Dexamethasone treatment. RESULTS: Innate expression of Notch pathway genes were significantly overexpressed in GTM cells with no discernible differences observed between HF/LF cells in either NTM or GTM cells cultured on plastic substrates. With 6 h of cyclical strain, a subset of Notch pathway genes presented with altered expression. Expression of Notch receptors/ligands/receptors/inhibitors progressively declined with increasing stiffness and this correlated with phagocytic ability of NTM cells. Dexamethasone treatment decreased phagocytosis regardless of stiffness or cells isolated from segmental outflow regions. CONCLUSIONS: We demonstrate here that the Notch expression in cultured TM cells differ intrinsically between GTM vs NTM, and by substratum cues (cyclical strain and stiffness). Of import, the most apparent differences in gene expression were observed as a function of substratum stiffness which closely followed phagocytic ability of cells. Interestingly, on soft substrates (mimicking normal TM stiffness) Notch expression and phagocytosis was highest, while both expression and phagocytosis was significantly lower on stiffer substrates (mimicking glaucomatous stiffness) regardless of DEX treatment. Such context dependent changes suggest Notch pathway may play differing roles in disease vs homeostasis. Studies focused on understanding the mechanistic role of Notch (if any) in outflow homeostasis are thus warranted.


Subject(s)
Gene Expression Regulation/physiology , Glaucoma/metabolism , Receptors, Notch/genetics , Trabecular Meshwork/metabolism , Aged , Aged, 80 and over , Blotting, Western , Cells, Cultured , Dexamethasone/pharmacology , Female , Glaucoma/pathology , Glucocorticoids/pharmacology , Humans , Male , Mechanotransduction, Cellular , Middle Aged , Phagocytosis/physiology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Tissue Donors , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics , Transforming Growth Factor beta/genetics , Wnt Proteins/genetics , YAP-Signaling Proteins/genetics
13.
Exp Eye Res ; 214: 108888, 2022 01.
Article in English | MEDLINE | ID: mdl-34896106

ABSTRACT

The trabecular meshwork (TM) is the leading site of aqueous humor outflow in the eye and plays a critical role in maintaining normal intraocular pressure. When the TM fails to maintain normal intraocular pressure, glaucoma may develop. Mitochondrial damage has previously been found in glaucomatous TM cells; however, the precise metabolic activity of glaucomatous TM cells has yet to be quantitatively assessed. Using dexamethasone (Dex) treated primary human TM cells to model glaucomatous TM cells, we measure the respiratory and glycolytic activity of Dex-treated TM cells with an extracellular flux assay. We found that Dex-treated TM cells had quantifiably altered metabolic profiles, including increased spare respiratory capacity and ATP production rate from oxidative phosphorylation. Therefore, we propose that reversing or preventing these metabolic changes may represent an avenue for future research.


Subject(s)
Adenosine Triphosphate/metabolism , Cytoskeletal Proteins/metabolism , Dexamethasone/pharmacology , Eye Proteins/metabolism , Glucocorticoids/pharmacology , Glycoproteins/metabolism , Trabecular Meshwork/drug effects , Aged , Cells, Cultured , Female , Humans , Male , Middle Aged , Tissue Donors , Trabecular Meshwork/cytology , Trabecular Meshwork/metabolism
14.
Biochim Biophys Acta Mol Basis Dis ; 1868(3): 166325, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34921975

ABSTRACT

OBJECTIVES: Glaucoma is characterized by progressive damage of the retinal ganglion cells (RGCs), resulting in irreversible vision loss. Cannabinoids (CBs) ameliorate several factors that contribute to the progression of glaucoma, including increased intraocular pressure (IOP), degeneration of RGC and optical nerve (ON) damage. However, a direct correlation of specific CBs with the molecular events pertaining to glaucoma pathology is not well established. Therefore, this study aims to evaluate the role of cannabinol (CBN) on RGC protection, modulation of IOP, and its effects on the level of extracellular matrix (ECM) proteins using both in vitro and in vivo models of glaucoma. METHODS AND RESULTS: When exposed to elevated hydrostatic pressure, CBN, in a dose-dependent manner, protected differentiated mouse 661W retinal ganglion precursor-like cells from pressure-induced toxicity. In human trabecular meshwork cells (hTM), CBN attenuated changes in the ECM proteins, including fibronectin and α-smooth muscle actin (α-SMA), as well as mitogen-activated protein kinases (phospho-ERK1/2) in the presence or absence of transforming growth factor-beta 2 (TGF-ß2) induced stress. Ocular pharmacokinetic parameters were evaluated post-intravitreal (IVT) CBN delivery in vivo. Furthermore, we demonstrated that IVT-administered CBN improved pattern electroretinogram (pERG) amplitudes and reduced IOP in a rat episcleral vein laser photocoagulation model of glaucoma. CONCLUSION: CBN promotes neuroprotection, abrogates changes in ECM protein, and normalizes the IOP levels in the eye. Therefore, our observations in the present study indicate a therapeutic potential for CBN in the treatment of glaucoma.


Subject(s)
Cannabinol/pharmacology , Disease Models, Animal , Glaucoma/drug therapy , Intraocular Pressure/drug effects , Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/drug effects , Trabecular Meshwork/drug effects , Animals , Glaucoma/metabolism , Glaucoma/pathology , Male , Mice , Rats , Rats, Wistar , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Trabecular Meshwork/metabolism , Trabecular Meshwork/pathology , Transforming Growth Factor beta2/metabolism
15.
Int J Mol Sci ; 22(21)2021 Nov 07.
Article in English | MEDLINE | ID: mdl-34769470

ABSTRACT

PURPOSE: The objective of the current study was to evaluate the effects of the autotaxin (ATX)-lysophosphatidic acid (LPA) signaling axis on the human trabecular meshwork (HTM) in two-dimensional (2D) and three-dimensional (3D) cultures of HTM cells. METHODS: The effects were characterized by transendothelial electrical resistance (TEER) and FITC-dextran permeability (2D), measurements of size and stiffness (3D), and the expression of several genes, including extracellular matrix (ECM) molecules, their modulators, and endoplasmic reticulum (ER) stress-related factors. RESULTS: A one-day exposure to 200 nM LPA induced significant down-sizing effects of the 3D HTM spheroids, and these effects were enhanced slightly on longer exposure. The TEER and FITC-dextran permeability data indicate that LPA induced an increase in the barrier function of the 2D HTM monolayers. A one-day exposure to a 2 mg/L solution of ATX also resulted in a significant decrease in the sizes of the 3D HTM spheroids, and an increase in stiffness was also observed. The gene expression of several ECMs, their regulators and ER-stress related factors by the 3D HTM spheroids were altered by both ATX and LPA, but in different manners. CONCLUSIONS: The findings presented herein suggest that ATX may have additional roles in the human TM, in addition to the ATX-LPA signaling axis.


Subject(s)
Lysophospholipids/pharmacology , Phosphoric Diester Hydrolases/pharmacology , Trabecular Meshwork/drug effects , Cell Culture Techniques/methods , Cells, Cultured , Humans , Phosphoric Diester Hydrolases/physiology , Spheroids, Cellular/drug effects , Spheroids, Cellular/physiology , Trabecular Meshwork/physiology
16.
Molecules ; 26(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34770791

ABSTRACT

Effects of a pan-ROCK-inhibitor, ripasudil (Rip), and a ROCK2 inhibitor, KD025 on dexamethasone (DEX)-treated human trabecular meshwork (HTM) cells as a model of steroid-induced glaucoma were investigated. In the presence of Rip or KD025, DEX-treated HTM cells were subjected to permeability analysis of 2D monolayer by transendothelial electrical resistance (TEER) and FITC-dextran permeability, physical properties, size and stiffness analysis (3D), and qPCR of extracellular matrix (ECM), and their modulators. DEX resulted in a significant increase in the permeability, as well as a large and stiff 3D spheroid, and those effects were inhibited by Rip. In contrast, KD025 exerted opposite effects on the physical properties (down-sizing and softening). Furthermore, DEX induced several changes of gene expressions of ECM and their modulators were also modulated differently by Rip and KD025. The present findings indicate that Rip and KD025 induced opposite effects toward 2D and 3D cell cultures of DEX-treated HTM cells.


Subject(s)
Dexamethasone/pharmacology , Protein Kinase Inhibitors/pharmacology , Trabecular Meshwork/cytology , Trabecular Meshwork/drug effects , rho-Associated Kinases/antagonists & inhibitors , Biomarkers , Cell Culture Techniques , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Humans
17.
Int J Mol Sci ; 22(22)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34830007

ABSTRACT

Primary Open-Angle Glaucoma (POAG) is a neurodegenerative disease, and its clinical outcomes lead to visual field constriction and blindness. POAG's etiology is very complex and its pathogenesis is mainly explained through both mechanical and vascular theories. The trabecular meshwork (TM), the most sensitive tissue of the eye anterior segment to oxidative stress (OS), is the main tissue involved in early-stage POAG, characterized by an increase in pressure. Preclinical assessments of neuroprotective drugs on animal models have not always shown correspondence with human clinical studies. In addition, intra-ocular pressure management after a glaucoma diagnosis does not always prevent blindness. Recently, we have been developing an innovative in vitro 3Dadvanced human trabecular cell model on a millifluidicplatform as a tool to improve glaucoma studies. Herein, we analyze the effects of prolonged increased pressure alone and, in association with OS, on such in vitro platform. Moreover, we verify whethersuch damaged TM triggers apoptosis on neuron-like cells. The preliminary results show that TM cells are less sensitive to pressure elevation than OS, and OS-damaging effects were worsened by the pressure increase. The stressed TM releases harmful signals, which increase apoptosis stimuli on neuron-like cells, suggesting its pivotal role in the glaucoma cascade.


Subject(s)
Glaucoma, Open-Angle/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Trabecular Meshwork/drug effects , Apoptosis/drug effects , Cell Line , Eye/metabolism , Eye/pathology , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/pathology , Humans , Intraocular Pressure/drug effects , Trabecular Meshwork/metabolism , Trabecular Meshwork/pathology
18.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34830390

ABSTRACT

Elevated intraocular pressure (IOP) is a major risk factor in developing primary open angle glaucoma (POAG), which is the most common form of glaucoma. Transforming growth factor-beta 2 (TGFß2) is a pro-fibrotic cytokine that plays an important role in POAG pathogenesis. TGFß2 induced extracellular matrix (ECM) production, deposition and endoplasmic reticulum (ER) stress in the trabecular meshwork (TM) contribute to increased aqueous humor (AH) outflow resistance and IOP elevation. Drugs which alter the glaucomatous fibrotic changes and ER stress in the TM may be effective in reducing ocular hypertension. Astragaloside IV (AS.IV), a novel saponin isolated from the roots of Astragalus membranaceus, has demonstrated antifibrotic and ER stress lowering effects in various tissues during disease conditions. However, the effect of AS.IV on glaucomatous TM fibrosis, ER stress and ocular hypertension has not been studied. Primary human TM cells treated with AS.IV decreased TGFß2 induced ECM (FN, Col-I) deposition and ER stress (KDEL, ATF4 and CHOP). Moreover, AS.IV treatment reduced TGFß2 induced NF-κB activation and αSMA expression in TM cells. We found that AS.IV treatment significantly increased levels of matrix metalloproteases (MMP9 and MMP2) and MMP2 enzymatic activity, indicating that the antifibrotic effects of AS.IV are mediated via inhibition of NF-κB and activation of MMPs. AS.IV treatment also reduced ER stress in TM3 cells stably expressing mutant myocilin. Interestingly, the topical ocular AS.IV eye drops (1 mM) significantly decreased TGFß2 induced ocular hypertension in mice, and this was associated with a decrease in FN, Col-1 (ECM), KDEL (ER stress) and αSMA in mouse TM tissues. Taken together, the results suggest that AS.IV prevents TGFß2 induced ocular hypertension by modulating ECM deposition and ER stress in the TM.


Subject(s)
Glaucoma, Open-Angle/drug therapy , Ocular Hypertension/drug therapy , Saponins/pharmacology , Transforming Growth Factor beta2/genetics , Triterpenes/pharmacology , Animals , Aqueous Humor/drug effects , Disease Models, Animal , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/pathology , Humans , Intraocular Pressure/drug effects , Mice , Ocular Hypertension/genetics , Ocular Hypertension/pathology , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology
19.
PLoS One ; 16(10): e0258911, 2021.
Article in English | MEDLINE | ID: mdl-34673834

ABSTRACT

Trabecular meshwork constitutes the conventional outflow pathway and controls intraocular pressure by regulating aqueous outflow. Mechanical stimulation has been studied as one of the triggers to regulate aqueous outflow in trabecular meshwork, but it is not well understood. We investigated that how transient receptor potential cation channel subfamily V member 4 (TRPV4) functions in human trabecular meshwork cells (HTMC) and affects intraocular pressure (IOP). HTMC were treated with TRPV4 siRNA, followed by incubation for 24 hours. We confirmed the suppression of TRPV4 mRNA expression and the reduction of Ca2+ influx by the TRPV4 agonist GSK1016790A in TRPV4 siRNA-treated HTMC. TRPV4 siRNA-treated HTMC exhibited a significant reduction in Ca2+ influx and production of arachidonic acid and prostaglandin (PG) E2 induced by mechanical stretch, and direct activation of TRPV4 by GSK1016790A increased production of arachidonic acid, PGE2, and PGD2 and inhibited gel contraction. Furthermore, TRPV4-deficient mice had higher IOP than wild-type mice, and GSK1016790A administration lowered IOP. These results suggest that TRPV4 mediates the cellular response induced by trabecular meshwork stretch, leading to IOP reduction through the production of prostaglandins and inhibition of cell contraction. Targeting TRPV4 may have therapeutic benefits that lead to lowering IOP in glaucoma patients.


Subject(s)
Arachidonic Acid/metabolism , Dinoprostone/metabolism , Intraocular Pressure/physiology , TRPV Cation Channels/metabolism , Trabecular Meshwork/metabolism , Animals , Humans , Intraocular Pressure/drug effects , Leucine/analogs & derivatives , Leucine/pharmacology , Mice , Mice, Knockout , Physical Stimulation , RNA, Small Interfering , Sulfonamides/pharmacology , TRPV Cation Channels/genetics , Trabecular Meshwork/drug effects
20.
Sci Rep ; 11(1): 18169, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34518638

ABSTRACT

Glaucoma treatment is usually initiated with topical medication that lowers the intraocular pressure (IOP) by reducing the aqueous production, enhancing the aqueous outflow, or both. However, the effect of topical IOP-lowering medications on the microstructures of the aqueous outflow pathway are relatively unknown. In this retrospective, observational study, 56 treatment-naïve patients with primary open-angle glaucoma were enrolled. Images of the nasal and temporal corneoscleral limbus were obtained using anterior segment optical coherence tomography (AS-OCT). The conjunctival vessels and iris anatomy were used as landmarks to select the same limbal area scan, and the trabecular meshwork (TM) width, TM thickness, and Schlemm's canal (SC) area were measured before and after using the IOP-lowering agents for 3 months. Among the 56 patients enrolled, 33 patients used prostaglandin (PG) analogues, and 23 patients used dorzolamide/timolol fixed combination (DTFC). After 3 months of DTFC usage, the TM width, TM thickness, and SC area did not show significant changes in either the nasal or temporal sectors. Conversely, after prostaglandin analog usage, the TM thickness significantly increased, and the SC area significantly decreased (all P < 0.01). These findings warrant a deeper investigation into their relationship to aqueous outflow through the conventional and unconventional outflow pathways after treatment with PG analogues.


Subject(s)
Intraocular Pressure , Prostaglandins, Synthetic/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Timolol/pharmacology , Trabecular Meshwork/pathology , Administration, Topical , Drug Combinations , Humans , Intraocular Pressure/drug effects , Middle Aged , Tomography, Optical Coherence , Trabecular Meshwork/diagnostic imaging , Trabecular Meshwork/drug effects , Trabecular Meshwork/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL