Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 154
1.
Invest Ophthalmol Vis Sci ; 65(5): 41, 2024 May 01.
Article En | MEDLINE | ID: mdl-38809543

Purpose: The rat controlled elevation of intraocular pressure (CEI) model allows study of in vivo responses to short-term exposure to defined intraocular pressures (IOP). In this study, we used NanoString technology to investigate in vivo IOP-related gene responses in the trabecular meshwork (TM) and optic nerve head (ONH) simultaneously from the same animals. Methods: Male and female rats (N = 35) were subjected to CEI for 8 hours at pressures simulating mean, daytime normotensive rat IOP (CEI-20), or 2.5× IOP (CEI-50). Naïve animals that received no anesthesia or surgical interventions served as controls. Immediately after CEI, TM and ONH tissues were dissected, RNA was isolated, and samples were analyzed with a NanoString panel containing 770 genes. Postprocessing, raw count data were uploaded to ROSALIND for differential gene expression analyses. Results: For the TM, 45 IOP-related genes were significant in the CEI-50 versus CEI-20 and CEI-50 versus naïve comparisons, with 15 genes common to both comparisons. Bioinformatics analysis identified Notch and transforming growth factor beta (TGFß) pathways to be the most up- and downregulated Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, respectively. For ONH, 22 significantly differentially regulated genes were identified in the CEI-50 versus naïve comparison. Pathway analysis identified defense response and immune response as two significantly upregulated biological process pathways. Conclusions: This study demonstrated the ability to assay short-term IOP-responsive genes in both TM and ONH tissues simultaneously. In the TM, downregulation of TGFß pathway genes suggests that TM responses may reduce TGFß-induced extracellular matrix synthesis. For ONH, the initial response to short-term elevated IOP may be protective.


Disease Models, Animal , Intraocular Pressure , Ocular Hypertension , Optic Disk , Trabecular Meshwork , Animals , Trabecular Meshwork/metabolism , Intraocular Pressure/physiology , Rats , Male , Female , Optic Disk/metabolism , Ocular Hypertension/genetics , Ocular Hypertension/physiopathology , Gene Expression Regulation/physiology , Gene Expression Profiling , Rats, Sprague-Dawley
2.
Cell Rep Med ; 5(5): 101524, 2024 May 21.
Article En | MEDLINE | ID: mdl-38670096

The carbonic anhydrase 2 (Car2) gene encodes the primary isoenzyme responsible for aqueous humor (AH) production and plays a major role in the regulation of intraocular pressure (IOP). The CRISPR-Cas9 system, based on the ShH10 adenovirus-associated virus, can efficiently disrupt the Car2 gene in the ciliary body. With a single intravitreal injection, Car2 knockout can significantly and sustainably reduce IOP in both normal mice and glaucoma models by inhibiting AH production. Furthermore, it effectively delays and even halts glaucomatous damage induced by prolonged high IOP in a chronic ocular hypertension model, surpassing the efficacy of clinically available carbonic anhydrase inhibitors such as brinzolamide. The clinical application of CRISPR-Cas9 based disruption of Car2 is an attractive therapeutic strategy that could bring additional benefits to patients with glaucoma.


CRISPR-Cas Systems , Carbonic Anhydrase II , Ciliary Body , Glaucoma , Intraocular Pressure , Animals , Glaucoma/genetics , Glaucoma/pathology , Glaucoma/metabolism , CRISPR-Cas Systems/genetics , Ciliary Body/metabolism , Ciliary Body/pathology , Carbonic Anhydrase II/genetics , Carbonic Anhydrase II/metabolism , Mice , Aqueous Humor/metabolism , Humans , Disease Models, Animal , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/therapeutic use , Gene Deletion , Mice, Inbred C57BL , Ocular Hypertension/genetics , Ocular Hypertension/pathology
3.
Mol Aspects Med ; 94: 101218, 2023 12.
Article En | MEDLINE | ID: mdl-37976898

More than 76 million people worldwide are afflicted with the neurodegenerative eye diseases described and grouped together as glaucoma. A common feature amongst the many forms of glaucoma is chronically elevated intraocular pressure (IOP) within the anterior chamber of the eye that physically damages the retina, optic nerve and parts of the brain connected with visual perception. The mediators of the contusing raised IOP responsible for such damage and loss of vision include locally released inflammatory agents, tissue remodeling enzymes and infiltrating immune cells which damage the retinal ganglion cell (RGC) axons and eventually kill a significant number of the RGCs. Additional culprits include genetic defects of the patient that involve aberrations in receptors, enzymes and/or endogenous ligands and possible over- or under-production of the latter. Other genetic abnormalities may include issues with signal transduction machinery within key cells of critical tissues in the front (e.g. trabecular meshwork [TM] and Schlemm's canal [SC]) and back of the eye (e.g. retinal ganglion cells and their axons). Genome-wide associated studies (GWAS) coupled with next generation sequencing have provided powerful linkage of certain gene defects and polymorphic variants to the onset and progression of diseases of the tissues involved in fluid dynamics in the TM and SC, and many retinal elements (lamina cribosa, optic nerve head) at the back of the eye which cause ocular hypertension (OHT) and glaucomatous optic neuropathy (GON), respectively. Despite the availability of some drugs, fluid drainage microshunts and full surgical techniques to lower and control intraocular pressure, the major modifiable biomarker of open-angle and other forms of glaucoma, their side-effect profiles, less than optimum effectiveness and short duration of action present opportunities to clinically manage the glaucomas with next generation of treatments with high therapeutic indices, including gene therapies. Thus, identification, characterization and deployment of genetic data coupled with traditional drug discovery and novel gene replacement, gene editing and genetic engineering technologies may provide some solutions to the aforementioned problems. These aspects will be discussed in this article.


Glaucoma , Ocular Hypertension , Animals , Humans , Disease Models, Animal , Glaucoma/therapy , Glaucoma/drug therapy , Ocular Hypertension/genetics , Ocular Hypertension/therapy , Intraocular Pressure , Retina
4.
Aging (Albany NY) ; 15(19): 10705-10731, 2023 10 10.
Article En | MEDLINE | ID: mdl-37819813

Glaucoma is a leading cause of irreversible vision loss characterized by retinal neurodegeneration. Circular RNAs (circRNAs) have emerged as the potential biomarkers and therapeutic targets for neurodegenerative diseases. However, the expression profiling of circRNAs in glaucomatous neurodegeneration has not been fully understood. In this study, we built a glaucomatous neurodegeneration model via the injection of microbeads into anterior chamber. circRNA expression profile and bioinformatics analysis revealed that compared with normal retinas, 171 circRNAs were dysregulated in the glaucomatous retinas, including 101 up-regulated circRNAs and 70 down-regulated circRNAs. Detecting the level of circular RNA-glycine receptor α2 subunit gene (cGlra2) in aqueous humor made it possible to distinguish glaucoma patients from cataract patients. Silencing of cGlra2 protected against oxidative stress- or hydrostatic pressure-induced retinal ganglion cell (RGC) injury in vitro. Moreover, silencing of cGlra2 retarded ocular hypertension-induced retinal neurodegeneration in vivo as shown by increased TUJ1 staining, reduced reactive gliosis, decreased retinal cell apoptosis, enhanced visual acuity, and improved retinal function. cGlra2 acted as a miRNA sponge to regulate RGC function through cGlra2/miR-144/BCL2L11 signaling axis. Collectively, this study provides novel insights into the underlying mechanism of retinal neurodegeneration and highlights the potential of cGlra2 as a target for the diagnosis and treatment of glaucoma.


Glaucoma , Ocular Hypertension , Humans , Animals , RNA, Circular/genetics , RNA, Circular/metabolism , Retina/metabolism , Ocular Hypertension/genetics , Ocular Hypertension/metabolism , Retinal Ganglion Cells , Disease Models, Animal
5.
Methods Mol Biol ; 2708: 77-97, 2023.
Article En | MEDLINE | ID: mdl-37558962

Viral transduction of the mouse trabecular meshwork using a variety of transgenes associated with glaucoma generates an inducible and reproducible method for generating ocular hypertension due to increased aqueous humor outflow resistance of the conventional outflow pathway. Both adenovirus serotype 5 (Ad5) and lentiviruses have selective tropism for the mouse trabecular meshwork with intraocular injections. Accurate intraocular pressures are easily measured using a rebound tonometer, and aqueous humor outflow facilities can be measured in anesthetized live mice.


Glaucoma , Ocular Hypertension , Mice , Animals , Ocular Hypertension/genetics , Ocular Hypertension/metabolism , Intraocular Pressure , Trabecular Meshwork/metabolism , Aqueous Humor/metabolism
6.
Neurotherapeutics ; 20(3): 896-907, 2023 Apr.
Article En | MEDLINE | ID: mdl-36941497

SIRT1 prevents retinal ganglion cell (RGC) loss in several acute and subacute optic neuropathy models following pharmacologic activation or genetic overexpression. We hypothesized that adeno-associated virus (AAV)-mediated overexpression of SIRT1 in RGCs in a chronic ocular hypertension model can reduce RGC loss, thereby preserving visual function by sustained therapeutic effect. A control vector AAV-eGFP and therapeutic vector AAV-SIRT1 were constructed and optimized for transduction efficiency. A magnetic microbead mouse model of ocular hypertension was optimized to induce a time-dependent and chronic loss of visual function and RGC degeneration. Mice received intravitreal injection of control or therapeutic AAV in which a codon-optimized human SIRT1 expression is driven by a RGC selective promoter. Intraocular pressure (IOP) was measured, and visual function was examined by optokinetic response (OKR) weekly for 49 days following microbead injection. Visual function, RGC survival, and axon numbers were compared among control and therapeutic AAV-treated animals. AAV-eGFP and AAV-SIRT1 showed transduction efficiency of ~ 40%. AAV-SIRT1 maintains the transduction of SIRT1 over time and is selectively expressed in RGCs. Intravitreal injections of AAV-SIRT1 in a glaucoma model preserved visual function, increased RGC survival, and reduced axonal degeneration compared with the control construct. Over-expression of SIRT1 through AAV-mediated gene transduction indicates a RGC-selective component of neuroprotection in multiple models of acute optic nerve degeneration. Results here show a neuroprotective effect of RGC-selective gene therapy in a chronic glaucoma model characterized by sustained elevation of IOP and subsequent RGC loss. Results suggest that this strategy may be an effective therapeutic approach for treating glaucoma, and warrants evaluation for the treatment of other chronic neurodegenerative diseases.


Glaucoma , Ocular Hypertension , Humans , Mice , Animals , Retinal Ganglion Cells/metabolism , Intraocular Pressure , Sirtuin 1/genetics , Sirtuin 1/metabolism , Glaucoma/genetics , Glaucoma/therapy , Ocular Hypertension/genetics , Ocular Hypertension/therapy , Genetic Therapy/methods , Disease Models, Animal , Axons/metabolism
7.
Int J Mol Sci ; 24(3)2023 Jan 20.
Article En | MEDLINE | ID: mdl-36768422

Analysis of the genotype that predicts the phenotypic characteristics of a cohort of glaucoma and ocular hypertension patients, and the correlation with their personal pharmacological response to beta-blockers (BB) and prostaglandin analogues (PGA). Prospective study that included 139 eyes from 72 patients under BB and/or PGA treatment, and in some cases other types of ocular hypotensive treatments. Five single-nucleotide polymorphisms were genotyped by real-time PCR assays: prostaglandin-F2α receptor (rs3766355, rs3753380); cytochrome-P450 2D6 (rs16947, rs769258); and beta-2-adrenergic receptor (rs1042714). Other studied variables were mean deviation (MD) of visual field, previous ocular interventions, medical treatment, baseline (bIOP), and treated intraocular pressure (tIOP). From a total of 139 eyes, 71 (51.1%) were left eyes. The main diagnosis was primary open angle glaucoma (66.2%). A total of 57 (41%) eyes were under three or more medications (PGA + BB + other) and, additionally, 57 eyes (41%) had had some kind of glaucoma surgery. The mean bIOP and tIOP were 26.55 ± 8.19 and 21.01 ± 5.54 mmHg, respectively. Significant differences in tIOP were found between heterozygous (HT) (21.07 ± 0.607 mmHg) and homozygous (HM) (20.98 ± 0.639 mmHg) rs3766355 with respect to wildtype individuals (16 ± 1.08 mmHg) (p = 0.031). The MD values presented significant differences between wildtype rs3766355 (-2 ± 2.2 dB), HT (-3.87 ± 4 dB), and HM carriers (-9.37 ± 9.51 dB) (p = 0.009). Significant differences were also observed between the MD in wildtype rs3753380 (-6.1 ± 8.67 dB), HT (-9.02 ± 8.63 dB), and HM carriers (-9.51 ± 7.44 dB) (p = 0.017). Patients carrying the variant rs3766355 in HM or HT presented clinically-significantly higher tIOP than wildtype patients. Additionally, some differences in MD were found in rs3766355 and rs3753380 carriers, and the more alleles that were affected, the worse the MD value, meaning greater severity of the glaucoma. Poor response to treatment and more visual field damage may be associated with being a carrier of these mutated alleles.


Glaucoma, Open-Angle , Glaucoma , Ocular Hypertension , Humans , Glaucoma, Open-Angle/drug therapy , Glaucoma, Open-Angle/genetics , Prospective Studies , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Glaucoma/drug therapy , Glaucoma/genetics , Ocular Hypertension/drug therapy , Ocular Hypertension/genetics , Intraocular Pressure , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Genotype , Phenotype , Prostaglandins, Synthetic/pharmacology , Prostaglandins, Synthetic/therapeutic use
8.
Mol Genet Genomics ; 298(1): 13-26, 2023 Jan.
Article En | MEDLINE | ID: mdl-36222912

Glucocorticosteroids commonly used to treat certain ocular inflammatory conditions cause an unwarranted elevation in intraocular pressure (IOP) leading to steroid-induced ocular hypertension (OHT). This study aims to identify novel genetic variants in the Indian population associated with steroid responsiveness, specifically to that of intravitreal Triamcinolone acetonide (TA) injections, which leads to OHT in 27% of the TA-treated Indian subjects. Genetic determinants and pathways regulating TA-OHT progression were investigated by applying whole-genome sequencing (WGS) on DNA extracted from 53 blood samples that included TA responders and non-responders. Sequencing analysis yielded 45 intronic and 49 exonic variants to be associated with TA-OHT, which are known to play a vital role in eye, heart, brain, and bone deformities. Of these, the most significant genetic variant associated with TA-OHT was further considered for molecular dynamics (MD) simulation studies. Variants in the CRPPA, PLOD1, ARHGAP1, TIMELESS and TNFSF4 genes were found to be directly implicating TA-OHT. Furthermore, these genes were enriched in pathways associated with cardiomyopathy, focal adhesion, extracellular matrix, and actin cytoskeleton reorganization. MD simulation studies revealed that the top significant variant (rs141625803) in the CRPPA gene possesses a high pathogenic and structurally destabilizing effect. Thus, novel genetic variants that could be significantly associated with the TA-OHT progression were identified in this study. Validation of these targets in a larger cohort of patients along with their functional analysis would inform on the disease, thereby adding to the existing knowledge on the pathophysiology of TA-OHT.


Ocular Hypertension , Triamcinolone Acetonide , Humans , Triamcinolone Acetonide/adverse effects , Glucocorticoids/adverse effects , Retrospective Studies , Ocular Hypertension/chemically induced , Ocular Hypertension/genetics , Intraocular Pressure , OX40 Ligand
9.
Transl Vis Sci Technol ; 11(11): 8, 2022 11 01.
Article En | MEDLINE | ID: mdl-36355386

Purpose: Ocular hypertension is a significant risk factor for vision loss in glaucoma caused by the death of retinal ganglion cells (RGCs). We investigated whether small heat shock proteins (sHsps) expressed in RGCs protect those cells against ocular hypertension in mice. Methods: AAV2 vectors encoding genes for one of the following four human sHsps: HSPB1, HSPB4, HSPB5, or HSPB6 were constructed for RGC-specific expression. Ischemia/reperfusion was induced by elevating the intraocular pressure (IOP) to 120 mm Hg for one hour, followed by a rapid return to normal IOP. Microbeads (MB) were injected into the anterior chamber of mice to induce ocular hypertension. RGC death and glial activation were assessed by immunostaining for Brn3a, RBPMS, Iba1, and glial fibrillary acid protein in retinal flat mounts. RGC axonal defects were evaluated by anterograde transport of intravitreally injected cholera toxin-B. RGC function was assessed by pattern electroretinography. Results: Among the sHsps, HspB1 offered the best protection against RGC death from ischemia/reperfusion injury in the mouse retina. Intravitreal administration of AAV2-HSPB1 either two weeks before or one week after instituting ocular hypertension resulted in significant prevention of RGC loss. The MB-injected mice showed RGC axonal transportation defects, but AAV2-HSPB1 administration significantly inhibited this defect. AAV2-HSPB1 prevented glial activation caused by ocular hypertension. More importantly, a single injection of AAV2-HSPB1 protected RGCs long-term in MB-injected eyes. Conclusions: The administration of AAV2-HSPB1 inhibited RGC death and axonal transport defects and reduced glial activation in a mouse model of ocular hypertension. Translational Relevance: Our results suggested that the intravitreal delivery of AAV2-HSPB1 could be developed as a gene therapy to prevent vision loss on a long-term basis in glaucoma patients.


Glaucoma , Ocular Hypertension , Humans , Mice , Animals , Retinal Ganglion Cells/metabolism , Axonal Transport , Ocular Hypertension/genetics , Ocular Hypertension/metabolism , Glaucoma/genetics , Glaucoma/prevention & control , Intraocular Pressure , Disease Models, Animal , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism
10.
Int J Mol Sci ; 23(13)2022 Jun 21.
Article En | MEDLINE | ID: mdl-35805889

Glaucoma is a multifactorial disease leading to irreversible blindness. Primary open-angle glaucoma (POAG) is the most common form and is associated with the elevation of intraocular pressure (IOP). Reduced aqueous humor (AH) outflow due to trabecular meshwork (TM) dysfunction is responsible for IOP elevation in POAG. Extracellular matrix (ECM) accumulation, actin cytoskeletal reorganization, and stiffening of the TM are associated with increased outflow resistance. Transforming growth factor (TGF) ß2, a profibrotic cytokine, is known to play an important role in the development of ocular hypertension (OHT) in POAG. An appropriate mouse model is critical in understanding the underlying molecular mechanism of TGFß2-induced OHT. To achieve this, TM can be targeted with recombinant viral vectors to express a gene of interest. Lentiviruses (LV) are known for their tropism towards TM with stable transgene expression and low immunogenicity. We, therefore, developed a novel mouse model of IOP elevation using LV gene transfer of active human TGFß2 in the TM. We developed an LV vector-encoding active hTGFß2C226,228S under the control of a cytomegalovirus (CMV) promoter. Adult C57BL/6J mice were injected intravitreally with LV expressing null or hTGFß2C226,228S. We observed a significant increase in IOP 3 weeks post-injection compared to control eyes with an average delta change of 3.3 mmHg. IOP stayed elevated up to 7 weeks post-injection, which correlated with a significant drop in the AH outflow facility (40.36%). Increased expression of active TGFß2 was observed in both AH and anterior segment samples of injected mice. The morphological assessment of the mouse TM region via hematoxylin and eosin (H&E) staining and direct ophthalmoscopy examination revealed no visible signs of inflammation or other ocular abnormalities in the injected eyes. Furthermore, transduction of primary human TM cells with LV_hTGFß2C226,228S exhibited alterations in actin cytoskeleton structures, including the formation of F-actin stress fibers and crossed-linked actin networks (CLANs), which are signature arrangements of actin cytoskeleton observed in the stiffer fibrotic-like TM. Our study demonstrated a mouse model of sustained IOP elevation via lentiviral gene delivery of active hTGFß2C226,228S that induces TM dysfunction and outflow resistance.


Glaucoma, Open-Angle , Ocular Hypertension , Actins/metabolism , Animals , Aqueous Humor/metabolism , Cells, Cultured , Disease Models, Animal , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/metabolism , Intraocular Pressure , Mice , Mice, Inbred C57BL , Ocular Hypertension/genetics , Ocular Hypertension/metabolism , Trabecular Meshwork/metabolism , Transforming Growth Factor beta2/metabolism
11.
J Neuroinflammation ; 18(1): 271, 2021 Nov 17.
Article En | MEDLINE | ID: mdl-34789280

BACKGROUND: Glaucoma, the leading cause of irreversible blindness worldwide, is a type of retinal disease characterized by the selective death of retinal ganglion cells (RGCs). However, the pathogenesis of glaucoma has not been fully elucidated. Transient receptor potential vanilloid 4 (TRPV4) is a pressure-sensitive and calcium-permeable cation channel. TRPV4 is widely distributed in the retina and its sustained activation leads to RGC death; indicating that TRPV4 may be a possible target for glaucoma treatment. Here, we investigated the effects of TRPV4 on RGC apoptosis in a rat model of chronic ocular hypertension (COH), then examined the mechanism underlying these effects. METHODS: The COH model was established by injection of micro-magnetic beads into the anterior chamber of adult male rats. The expression levels of TRPV4, glial fibrillary acidic protein, and inflammatory factors were assessed by immunohistochemistry and immunoblotting. RGC apoptosis and visual dysfunction were evaluated by TUNEL assay and photopic negative response. Functional expression of TRPV4 was examined by electrophysiology and calcium imaging. Real-time polymerase chain reaction and immunoblotting were employed to investigate the molecular mechanism underlying the effects of TRPV4 on tumor necrosis factor-α (TNF-α) release. RESULTS: We found that TRPV4 played an essential role in glaucoma, such that high levels of TRPV4 expression were associated with elevated intraocular pressure. Furthermore, TRPV4 activation was involved in glaucoma-induced RGC apoptosis and RGC-related reductions in visual function. Mechanistic investigation demonstrated that TRPV4 activation led to enhanced Müller cell gliosis and TNF-α release via the JAK2/STAT3/NF-kB pathway, while TRPV4 inhibition could reverse these effects. Finally, TRPV4 activation could lead to elevated expression of TNF receptor 1 in RGCs, while inhibition of TNF-α could reduce TRPV4-mediated RGC apoptosis. CONCLUSIONS: TRPV4 activation induces Müller cell gliosis and TNF-α elevation via the JAK2/STAT3/NF-κB pathway, which may exacerbate RGC apoptosis in glaucoma; these results suggest that TRPV4 can serve as a therapeutic target in glaucoma treatment.


Apoptosis/genetics , Ependymoglial Cells/pathology , Glaucoma/genetics , Janus Kinase 2/genetics , NF-kappa B/genetics , Retinal Ganglion Cells/pathology , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , TRPV Cation Channels/genetics , Tumor Necrosis Factor-alpha/metabolism , Animals , Glaucoma/pathology , Gliosis/pathology , Intraocular Pressure , Male , Ocular Hypertension/genetics , Ocular Hypertension/pathology , Rats , Rats, Wistar , Vision Disorders/genetics , Vision Disorders/pathology
12.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article En | MEDLINE | ID: mdl-34830387

Glaucoma is an optic neuropathy in which the degeneration of retinal ganglion cells (RGCs) results in irreversible vison loss. Therefore, neuroprotection of RGCs from glaucomatous afflictions is crucial for glaucoma treatment. In this study, we aimed to investigate the beneficial effects of statins in the protection of RGCs using a rat model. Glaucomatous injury was induced in rats by chronic ocular hypertension (OHT) achieved after performing a circumlimbal suture. The rats were given either statins such as simvastatin and atorvastatin or a solvent weekly for 6 weeks. Retina sections underwent hematoxylin and eosin, Brn3a, or cleaved casepase-3 staining to evaluate RGC survival. In addition, modulation of glial activation was assessed. While the retinas without statin treatment exhibited increased RGC death due to chronic OHT, statins promoted the survival of RGCs and reduced apoptosis. Statins also suppressed chronic OHT-mediated glial activation in the retina. Our results demonstrate that statins exert neuroprotective effects in rat retinas exposed to chronic OHT, which may support the prospect of statins being a glaucoma treatment.


Glaucoma/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Ocular Hypertension/drug therapy , Retinal Degeneration/drug therapy , Animals , Disease Models, Animal , Glaucoma/genetics , Glaucoma/pathology , Humans , Intraocular Pressure/drug effects , Neuroprotection/genetics , Neuroprotective Agents/pharmacology , Ocular Hypertension/genetics , Ocular Hypertension/pathology , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve Diseases/drug therapy , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Rats , Retina/drug effects , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Transcription Factor Brn-3A/chemistry , Transcription Factor Brn-3A/isolation & purification
13.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article En | MEDLINE | ID: mdl-34830390

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.


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
14.
Invest Ophthalmol Vis Sci ; 62(12): 7, 2021 09 02.
Article En | MEDLINE | ID: mdl-34499703

Purpose: Primary open angle glaucoma (POAG) is a leading cause of blindness worldwide with elevated intraocular pressure (IOP) as the most important risk factor. POAG IOP elevation is due to pathological changes in the trabecular meshwork (TM). Elevated TGFß2 contributes to these changes and increases IOP. We have shown that histone hyperacetylation is associated with TGFß2 elevation in the TM. In this study, we determined if clustered regularly interspaced short palindromic repeats (CRISPR) interference could specifically deacetylate histones and decrease TGFß2 in the TM. Methods: We tested the efficiency of different promoters in driving KRAB-dCAS9 expression in human TM cells. We also screened and determined the optimal sgRNA sequence in the inhibition of TGFß2. Chromatin immunoprecipitation-qPCR was used to determine the binding of KRAB-dCAS9. An adenovirus-mediated TGFß2-induced ocular hypertension (OHT) mouse model was used to determine the effect of the CRISPR interference system in vivo. Results: We found that the CRISPR interference system inhibited TGFß2 expression in human TM cells, and properly designed sgRNA targeted the promoter of the TGFß2 gene. Using sgRNA targeting the CMV promoter of the Ad5-CMV-TGFß2 viral vector, we found that lentivirus-mediated KRAB-dCAS9 and sgRNA expression was able to inhibit Ad5-CMV-TGFß2-induced OHT in C57BL/6J female and male mice eyes. This inhibition of OHT was associated with decreased levels of TGFß2 and extracellular matrix proteins in the mouse eye. Conclusions: Our results indicate that CRISPR interference is a useful tool for gene inhibition and may be a therapeutic approach to treat TGFß2-induced OHT.


Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Glaucoma, Open-Angle/genetics , Intraocular Pressure/physiology , Ocular Hypertension/genetics , Trabecular Meshwork/metabolism , Transforming Growth Factor beta2/adverse effects , Animals , Cells, Cultured , Disease Models, Animal , Female , Genetic Vectors , Glaucoma, Open-Angle/chemically induced , Glaucoma, Open-Angle/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Ocular Hypertension/chemically induced , Ocular Hypertension/metabolism , Signal Transduction , Trabecular Meshwork/pathology
15.
Cells ; 10(7)2021 06 22.
Article En | MEDLINE | ID: mdl-34206213

The purpose of this study was to characterize the miRNA profile of purified retinal ganglion cells (RGC) from healthy and diseased rat retina. Diseased retina includes those after a traumatic optic nerve crush (ONC), and after ocular hypertension/glaucoma. Rats were separated into four groups: healthy/intact, 7 days after laser-induced ocular hypertension, 2 days after traumatic ONC, and 7 days after ONC. RGC were purified from rat retina using microbeads conjugated to CD90.1/Thy1. RNA were sequenced using Next Generation Sequencing. Over 100 miRNA were identified that were significantly different in diseased retina compared to healthy retina. Considerable differences were seen in the miRNA expression of RGC 7 days after ONC, whereas after 2 days, few changes were seen. The miRNA profiles of RGC 7 days after ONC and 7 days after ocular hypertension were similar, but discrete miRNA differences were still seen. Candidate mRNA showing different levels of expression after retinal injury were manipulated in RGC cultures using mimics/AntagomiRs. Of the five candidate miRNA identified and subsequently tested for therapeutic efficacy, miR-194 inhibitor and miR-664-2 inhibitor elicited significant RGC neuroprotection, whereas miR-181a mimic and miR-181d-5p mimic elicited significant RGC neuritogenesis.


Glaucoma/genetics , Glaucoma/pathology , MicroRNAs/metabolism , Nerve Crush , Optic Nerve/pathology , Retinal Ganglion Cells/metabolism , Animals , Female , Gene Expression Regulation , Intraocular Pressure/genetics , Mice, Inbred C57BL , MicroRNAs/genetics , Neurites/metabolism , Neurogenesis/genetics , Neuroprotection/genetics , Ocular Hypertension/genetics , Ocular Hypertension/physiopathology , Rats, Sprague-Dawley
16.
JAMA Ophthalmol ; 139(7): 762-768, 2021 Jul 01.
Article En | MEDLINE | ID: mdl-34081096

IMPORTANCE: The c.1102C>T, p.(Gln368Ter) variant in the myocilin (MYOC) gene is a known risk allele for glaucoma. It is the most common MYOC risk variant for glaucoma among individuals of European ancestry, and its prevalence is highest in Finland. Furthermore, exfoliation syndrome has high prevalence in Scandinavia, making the Finnish population ideal to study the association of the variant with different types of glaucoma. OBJECTIVES: To examine the association and penetrance of MYOC p.(Gln368Ter) (rs74315329) variant with different types of glaucoma in a Finnish population. DESIGN, SETTING, AND PARTICIPANTS: This genetic association study included individuals of Finnish ancestry in the FinnGen project. The participants were collected from Finnish biobanks, and the disease end points were defined using nationwide registries. The MYOC c.1102C>T variant was either directly genotyped or imputed with microarrays. Recruitment of samples to FinnGen was initiated in 2017, and data analysis was performed between December 2019 and May 2020. MAIN OUTCOMES AND MEASURES: The main outcomes were odds ratios (ORs) and penetrance with different types of glaucoma and in different age groups. RESULTS: A total of 218 792 individuals were included in this study (mean [SD] age 52.4 [17.5] years; 123 579 women [56.5%]), including 8591 (3.9%) with glaucoma, 3412 (1.6%) with primary open-angle glaucoma, 1515 (0.7%) with exfoliation glaucoma, 892 (0.4%) with normal-tension glaucoma, and 4766 (2.2%) with suspected glaucoma. The minor allele frequency of MYOC p.(Gln368Ter) was 0.28%. Individuals with the heterozygous variant had higher odds of primary open-angle glaucoma (OR, 3.36; 95% CI, 2.55-4.37), overall glaucoma (OR, 2.58; 95% CI, 2.12-3.13), suspected glaucoma (OR, 2.53; 95% CI, 1.93-3.26), exfoliation glaucoma (OR, 2.61; 95% CI, 1.60-4.02), and undergoing glaucoma-related operations (OR, 5.45; 95% CI, 2.95-9.28). The penetrance of heterozygous MYOC p.(Gln368Ter) was 5.2% in individuals with primary open-angle glaucoma, 9.6% in individuals with glaucoma, 5.4% in individuals with suspected glaucoma, and 1.9% in individuals with exfoliation glaucoma. There was no significant association with normal-tension glaucoma (OR, 1.69; 95% CI, 0.72-3.35). CONCLUSIONS AND RELEVANCE: This genetic association study found that the MYOC p.(Gln368Ter) variant was associated with exfoliation glaucoma. The association with normal-tension glaucoma could not be replicated. These findings suggest that MYOC p.(Gln368Ter) was associated with open-angle glaucoma and exfoliation glaucoma in a Finnish population.


Exfoliation Syndrome , Glaucoma, Open-Angle , Glaucoma , Low Tension Glaucoma , Ocular Hypertension , Cytoskeletal Proteins , Exfoliation Syndrome/diagnosis , Exfoliation Syndrome/epidemiology , Exfoliation Syndrome/genetics , Eye Proteins/genetics , Female , Finland/epidemiology , Glaucoma/diagnosis , Glaucoma/epidemiology , Glaucoma/genetics , Glaucoma, Open-Angle/diagnosis , Glaucoma, Open-Angle/epidemiology , Glaucoma, Open-Angle/genetics , Glycoproteins , Humans , Intraocular Pressure , Male , Middle Aged , Mutation , Ocular Hypertension/genetics
17.
Mol Vis ; 27: 61-77, 2021.
Article En | MEDLINE | ID: mdl-33633440

PURPOSE: The effects of aqueous mediators possibly increasing the outflow resistance, transforming growth factor-ß1 (TGF-ß1), TGF-ß2, autotaxin (ATX), and lysophosphatidic acid (LPA) on human trabecular meshwork (hTM) cells and monkey Schlemm's canal endothelial (SCE) cells were characterized and compared, and the effects of intracameral application of these mediators on intraocular (IOP) elevation were also examined. METHODS: Cells were treated with TGF-ß1, TGF-ß2, ATX, LPA, or vehicle, and mRNA and protein expression levels of α-SMA, COL1A1, fibronectin, ß-catenin, and ZO-1 were examined with real-time quantitative PCR (RT-qPCR) or immunofluorescence analyses or both. The permeability of cell monolayers was measured by determining the transendothelial electrical resistance (TEER) or with the fluorescein isothiocyanate (FITC)-dextran permeability assay. IOP was evaluated in rabbit eyes after intracameral administration of the mediators. RESULTS: All mediators induced upregulation of α-SMA, COL1A1, and fibronectin in hTM cells. The effect of TGF-ß2 on mRNA expression of fibrotic markers was statistically significantly greater than that of TGF-ß1. The effects of ATX and LPA indicated the time-dependent difference in the upregulation of α-SMA, COL1A1, and fibronectin. The TEER and FITC-dextran permeability of the SCE cells was evaluated after treatment with TGF-ß1 and TGF-ß2, but no statistically significant change was observed within 24 h. ATX and LPA also reduced permeability statistically significantly after 3 h and 0.5 h, respectively, and the effect of LPA was more rapid compared to that of ATX. Statistically significant IOP elevation was observed in rabbit eyes as early as 0.5-2.0 h after ATX and LPA treatment and at 24 h after treatment with TGF-ß2. CONCLUSIONS: TGF-ß2 and ATX and LPA regulate aqueous outflow by modulation of hTM cells and SCE cells, and differences in timing between the effects of each mediator were observed. ATX and LPA showed more rapid effects on IOP elevation than TGF-ß2. It was suggested that TGF-ß2 and ATX/LPA are involved in increases of IOP, but the timing and sustainability differ between mediators, and they may play specific roles in different glaucoma subtypes.


Aqueous Humor/physiology , Intraocular Pressure/drug effects , Lysophospholipids/pharmacology , Ocular Hypertension/chemically induced , Phosphoric Diester Hydrolases/pharmacology , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta2/pharmacology , Actins/genetics , Actins/metabolism , Administration, Ophthalmic , Animals , Blotting, Western , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fibronectins/genetics , Fibronectins/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Macaca fascicularis , Middle Aged , Ocular Hypertension/genetics , Ocular Hypertension/metabolism , Ophthalmic Solutions , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
18.
Redox Biol ; 42: 101883, 2021 06.
Article En | MEDLINE | ID: mdl-33579667

Oxidative stress contributes to degeneration of retinal ganglion cells and their axons in glaucoma, a leading cause of irreversible blindness worldwide, through sensitivity to intraocular pressure (IOP). Here, we investigated early elevations in reactive oxygen species (ROS) and a role for the NRF2-KEAP1-ARE endogenous antioxidant response pathway using microbead occlusion to elevate IOP in mice. ROS levels peaked in the retina at 1- and 2-wks following IOP elevation and remained elevated out to 5-wks. Phosphorylation of NRF2 and antioxidant gene transcription and protein levels increased concomitantly at 2-wks after IOP elevation, along with phosphorylation of PI3K and AKT. Inhibiting PI3K or AKT signaling prevented NRF2 phosphorylation and reduced transcription of antioxidant-regulated genes. Ocular hypertensive mice lacking Nrf2 had elevated ROS and a diminished increase in antioxidant gene expression. They also exhibited earlier axon degeneration and loss of visual function. In conclusion, the NRF2-KEAP1-ARE pathway is endogenously activated early in ocular hypertension due to phosphorylation of NRF2 by the PI3K/AKT pathway and serves to slow the onset of axon degeneration and vision loss in glaucoma. These data suggest that exogenous activation of this pathway might further slow glaucomatous neurodegeneration.


Glaucoma , Ocular Hypertension , Animals , Disease Models, Animal , Glaucoma/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Ocular Hypertension/genetics , Oxidative Stress , Phosphatidylinositol 3-Kinases/genetics , Retina/metabolism
19.
Sci Rep ; 11(1): 1583, 2021 01 15.
Article En | MEDLINE | ID: mdl-33452295

Predicting the therapeutic response to ocular hypotensive drugs is crucial for the clinical treatment and management of glaucoma. Our aim was to identify a possible genetic contribution to the response to current pharmacological treatments of choice in a white Mediterranean population with primary open-angle glaucoma (POAG) or ocular hypertension (OH). We conducted a prospective, controlled, randomized, partial crossover study that included 151 patients of both genders, aged 18 years and older, diagnosed with and requiring pharmacological treatment for POAG or OH in one or both eyes. We sought to identify copy number variants (CNVs) associated with differences in pharmacological response, using a DNA pooling strategy of carefully phenotyped treatment responders and non-responders, treated for a minimum of 6 weeks with a beta-blocker (timolol maleate) and/or prostaglandin analog (latanoprost). Diurnal intraocular pressure reduction and comparative genome wide CNVs were analyzed. Our finding that copy number alleles of an intronic portion of the MLIP gene is a predictor of pharmacological response to beta blockers and prostaglandin analogs could be used as a biomarker to guide first-tier POAG and OH treatment. Our finding improves understanding of the genetic factors modulating pharmacological response in POAG and OH, and represents an important contribution to the establishment of a personalized approach to the treatment of glaucoma.


Co-Repressor Proteins/genetics , Glaucoma, Open-Angle/pathology , Ocular Hypertension/pathology , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Adult , Aged , Alleles , Biomarkers/metabolism , Cross-Over Studies , DNA Copy Number Variations , Female , Genome-Wide Association Study , Genotype , Glaucoma, Open-Angle/drug therapy , Glaucoma, Open-Angle/genetics , Humans , Intraocular Pressure/drug effects , Latanoprost/pharmacology , Latanoprost/therapeutic use , Male , Middle Aged , Ocular Hypertension/drug therapy , Ocular Hypertension/genetics , Prospective Studies , Timolol/pharmacology , Timolol/therapeutic use
20.
Cell Death Dis ; 11(9): 734, 2020 09 10.
Article En | MEDLINE | ID: mdl-32913260

Autophagy has a fundamental role in maintaining cell homeostasis. Although autophagy has been implicated in glaucomatous pathology, how it regulates retinal ganglion cell (RGC) injury is largely unknown. In the present work, we found that biphasic autophagy in RGCs occurred in a mouse model of chronic ocular hypertension (COH), accompanied by activation of Rac1, a member of the Rho family. Rac1 conditional knockout (Rac1 cKO) in RGCs attenuated RGC apoptosis, in addition to blocking the increase in the number of autophagosomes and the expression of autophagy-related proteins (Beclin1, LC3-II/I, and p62) in COH retinas. Electron micrograph and double immunostaining of LAMP1 and LC3B showed that Rac1 cKO accelerated autolysosome fusion in RGC axons of COH mice. Inhibiting the first autophagic peak with 3-methyladenine or Atg13 siRNA reduced RGC apoptosis, whereas inhibiting the second autophagic peak with 3-MA or blocking autophagic flux by chloroquine increased RGC apoptosis. Furthermore, Rac1 cKO reduced the number of autophagosomes and apoptotic RGCs induced by rapamycin injected intravitreally, which suggests that Rac1 negatively regulates mTOR activity. Moreover, Rac1 deletion decreased Bak expression and did not interfere with the interaction of Beclin1 and Bcl-2 or Bak in COH retinas. In conclusion, autophagy promotes RGC apoptosis in the early stages of glaucoma and results in autophagic cell death in later stages. Rac1 deletion alleviates RGC damage by regulating the cross talk between autophagy and apoptosis through mTOR/Beclin1-Bak. Interfering with the Rac1/mTOR signaling pathway may provide a new strategy for treating glaucoma.


Ocular Hypertension/genetics , Peptide Fragments/metabolism , Retinal Ganglion Cells/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Apoptosis , Cell Differentiation , Chronic Disease , Disease Models, Animal , Humans , Male , Mice , Ocular Hypertension/pathology
...