Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 294
Filter
1.
Invest Ophthalmol Vis Sci ; 65(5): 15, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38717426

ABSTRACT

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


Subject(s)
Collagen Type IV , Glaucoma , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction , Transforming Growth Factor beta , Animals , Mice , Anterior Eye Segment/metabolism , Anterior Eye Segment/pathology , Collagen Type IV/metabolism , Collagen Type IV/genetics , Disease Models, Animal , Glaucoma/metabolism , Glaucoma/genetics , Glaucoma/pathology , Intraocular Pressure/physiology , Mice, Inbred C57BL , Mutation , Optic Nerve/pathology , Optic Nerve/metabolism , Optic Nerve Diseases/metabolism , Optic Nerve Diseases/genetics , Phenotype , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Signal Transduction/physiology , Slit Lamp Microscopy , Tomography, Optical Coherence , Tonometry, Ocular , Transforming Growth Factor beta/metabolism
2.
Adv Biol (Weinh) ; 8(5): e2400018, 2024 05.
Article in English | MEDLINE | ID: mdl-38640945

ABSTRACT

Ophthalmic diseases affect many people, causing partial or total loss of vision and a reduced quality of life. The anterior segment of the eye accounts for nearly half of all visual impairment that can lead to blindness. Therefore, there is a growing demand for ocular research and regenerative medicine that specifically targets the anterior segment to improve vision quality. This study aims to generate a microfluidic platform for investigating the formation of the anterior segment of the eye derived from human induced pluripotent stem cells (hiPSC) under various spatial-mechanoresponsive conditions. Microfluidic platforms are developed to examine the effects of dynamic conditions on the generation of hiPSCs-derived ocular organoids. The differentiation protocol is validated, and mechanoresponsive genes are identified through transcriptomic analysis. Several culture strategies is implemented for the anterior segment of eye cells in a microfluidic chip. hiPSC-derived cells showed anterior eye cell characteristics in mRNA and protein expression levels under dynamic culture conditions. The expression levels of yes-associated protein and transcriptional coactivator PDZ binding motif (YAP/TAZ) and PIEZO1, varied depending on the differentiation and growth conditions of the cells, as well as the metabolomic profiles under dynamic culture conditions.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Anterior Eye Segment/cytology , Anterior Eye Segment/metabolism , Microfluidics/methods , Microfluidics/instrumentation , Organoids/metabolism , Organoids/cytology , YAP-Signaling Proteins/metabolism , Lab-On-A-Chip Devices , Transcription Factors/metabolism , Transcription Factors/genetics , Ion Channels/genetics , Ion Channels/metabolism
3.
JCI Insight ; 9(9)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38592784

ABSTRACT

Recent studies have uncovered that noncoding sequence variants may relate to Axenfeld-Rieger syndrome (ARS), a rare developmental anomaly with genetic heterogeneity. However, how these genomic regions are functionally and structurally associated with ARS is still unclear. In this study, we performed genome-wide linkage analysis and whole-genome sequencing in a Chinese family with ARS and identified a heterozygous deletion of about 570 kb (termed LOH-1) in the intergenic sequence between paired-like homeodomain transcription factor 2 (PITX2) and family with sequence similarity 241 member A. Knockout of LOH-1 homologous sequences caused ARS phenotypes in mice. RNA-Seq and real-time quantitative PCR revealed a significant reduction in Pitx2 gene expression in LOH-1-/- mice, while forkhead box C1 expression remained unchanged. ChIP-Seq and bioinformatics analysis identified a potential enhancer region (LOH-E1) within LOH-1. Deletion of LOH-E1 led to a substantial downregulation of the PITX2 gene. Mechanistically, we found a sequence (hg38 chr4:111,399,594-111,399,691) that is on LOH-E1 could regulate PITX2 by binding to RAD21, a critical component of the cohesin complex. Knockdown of RAD21 resulted in reduced PITX2 expression. Collectively, our findings indicate that a potential enhancer sequence that is within LOH-1 may regulate PITX2 expression remotely through cohesin-mediated loop domains, leading to ARS when absent.


Subject(s)
Anterior Eye Segment , Eye Abnormalities , Eye Diseases, Hereditary , Homeobox Protein PITX2 , Homeodomain Proteins , Transcription Factors , Animals , Female , Humans , Male , Mice , Anterior Eye Segment/abnormalities , Anterior Eye Segment/metabolism , DNA, Intergenic/genetics , Enhancer Elements, Genetic/genetics , Eye Abnormalities/genetics , Eye Diseases, Hereditary/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice, Knockout , Pedigree , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Exp Eye Res ; 234: 109603, 2023 09.
Article in English | MEDLINE | ID: mdl-37495069

ABSTRACT

Anterior segment dysgenesis is a severe developmental eye disorder that leads to blindness in children. The exact mechanisms underlying this condition remain elusive. Recently, an increasing amount of studies have focused on genes and signal transduction pathways that affect anterior segment dysgenesis;these factors include transcription factors, developmental regulators, extracellular matrix genes, membrane-related proteins, cytoskeleton proteins and other associated genes. To date, dozens of gene variants have been found to cause anterior segment dysgenesis. However, there is still a lack of effective treatments. With a broader and deeper understanding of the molecular mechanisms underlying anterior segment development in the future, gene editing technology and stem cell technology may be new treatments for anterior segment dysgenesis. Further studies on the mechanisms of how different genes influence the onset and progression of anterior segment dysgenesis are still needed.


Subject(s)
Anterior Eye Segment , Eye Abnormalities , Child , Humans , Anterior Eye Segment/metabolism , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Transcription Factors/genetics , Molecular Biology
5.
Proc Natl Acad Sci U S A ; 119(29): e2200914119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858321

ABSTRACT

The anterior segment of the eye consists of the cornea, iris, ciliary body, crystalline lens, and aqueous humor outflow pathways. Together, these tissues are essential for the proper functioning of the eye. Disorders of vision have been ascribed to defects in all of them; some disorders, including glaucoma and cataract, are among the most prevalent causes of blindness in the world. To characterize the cell types that compose these tissues, we generated an anterior segment cell atlas of the human eye using high-throughput single-nucleus RNA sequencing (snRNAseq). We profiled 195,248 nuclei from nondiseased anterior segment tissues of six human donors, identifying >60 cell types. Many of these cell types were discrete, whereas others, especially in the lens and cornea, formed continua corresponding to known developmental transitions that persist in adulthood. Having profiled each tissue separately, we performed an integrated analysis of the entire anterior segment, revealing that some cell types are unique to a single structure, whereas others are shared across tissues. The integrated cell atlas was then used to investigate cell type-specific expression patterns of more than 900 human ocular disease genes identified through either Mendelian inheritance patterns or genome-wide association studies.


Subject(s)
Anterior Eye Segment , Eye Diseases , Adult , Anterior Eye Segment/cytology , Anterior Eye Segment/metabolism , Aqueous Humor/cytology , Aqueous Humor/metabolism , Atlases as Topic , Ciliary Body/cytology , Ciliary Body/metabolism , Eye Diseases/genetics , Genome-Wide Association Study , Humans , Iris/cytology , Organ Specificity
6.
Invest Ophthalmol Vis Sci ; 63(1): 34, 2022 01 03.
Article in English | MEDLINE | ID: mdl-35077549

ABSTRACT

Purpose: To explore whether oxidative stress and premature senescence occur in the anterior segment of acute primary angle-closure (APAC) eyes after increased intraocular pressure. Methods: The eye samples of 21 APAC patients, 22 age-related cataract patients, and 10 healthy donors were included. Aqueous humor (AqH), iris, and anterior lens capsule samples were collected. The levels of oxidative stress markers and senescence-associated secretory phenotype (SASP)-related cytokines in AqH were estimated using relevant reagent kits and multiplex bead immunoassay technique. The intensity of relevant markers in anterior segment tissues was examined by immunofluorescence- and senescence-associated ß-galactosidase (SA-ß-gal) staining. Results: Oxidative stress marker levels elevated significantly in the AqH of APAC eyes. Reactive oxygen species (ROS) and 8-hydroxydeoxyguanosine levels were positively correlated with preoperative peak intraocular pressure and age, whereas reduced glutathione/oxidized glutathione (GSH/GSSH) ratio was negatively correlated with both parameters. The levels of several SASP-related cytokines were markedly increased. ROS and malondialdehyde levels were positively correlated with the levels of some SASP-related cytokines, whereas superoxide dismutase level and GSH/GSSH ratio showed an opposite trend. The number of cells positive for oxidative mitochondrial DNA damage and apoptosis-related markers increased in the iris and anterior lens capsule of the APAC group. Senescence-associated markers (p16, p21, and p53) and SA-ß-gal activity were increased in the iris of the APAC group. Conclusions: Oxidative stress and premature senescence occurred in the anterior segment of APAC patients, suggesting that they may be involved in the development of pathological changes in the anterior segment of APAC eyes.


Subject(s)
Anterior Eye Segment/metabolism , Cellular Senescence/physiology , Glaucoma, Angle-Closure/metabolism , Intraocular Pressure/physiology , Oxidative Stress , Tomography, Optical Coherence/methods , Acute Disease , Anterior Eye Segment/pathology , Female , Glaucoma, Angle-Closure/pathology , Humans , Male , Middle Aged , Prospective Studies , Retrospective Studies
7.
Int J Mol Sci ; 22(22)2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34830247

ABSTRACT

Topical drug delivery is one of the most challenging aspects of eye therapy. Eye drops are the most prevalent drug form, especially for widely distributed anterior segment eye diseases (cataracts, glaucoma, dry eye syndrome, inflammatory diseases, etc.), because they are convenient and easy to apply by patients. However, conventional drug formulations are usually characterized by short retention time in the tear film, insufficient contact with epithelium, fast elimination, and difficulties in overcoming ocular tissue barriers. Not more than 5% of the total drug dose administered in eye drops reaches the interior ocular tissues. To overcome the ocular drug delivery barriers and improve drug bioavailability, various conventional and novel drug delivery systems have been developed. Among these, nanosize carriers are the most attractive. The review is focused on the different drug carriers, such as synthetic and natural polymers, as well as inorganic carriers, with special attention to nanoparticles and nanomicelles. Studies in vitro and in vivo have demonstrated that new formulations could help to improve the bioavailability of the drugs, provide sustained drug release, enhance and prolong their therapeutic action. Promising results were obtained with drug-loaded nanoparticles included in in situ gel.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Drug Carriers/pharmacokinetics , Nanotechnology/methods , Ophthalmic Solutions/administration & dosage , Polymers/pharmacokinetics , Administration, Ophthalmic , Animals , Anterior Eye Segment/drug effects , Anterior Eye Segment/metabolism , Anterior Eye Segment/pathology , Anti-Inflammatory Agents/pharmacokinetics , Biological Availability , Cataract/drug therapy , Cataract/metabolism , Cataract/pathology , Drug Carriers/chemical synthesis , Drug Carriers/classification , Drug Liberation , Dry Eye Syndromes/drug therapy , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Glaucoma/drug therapy , Glaucoma/metabolism , Glaucoma/pathology , Humans , Micelles , Nanogels/chemistry , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Nanotechnology/instrumentation , Ophthalmic Solutions/pharmacokinetics , Polymers/chemical synthesis , Polymers/classification
8.
Development ; 148(16)2021 08 15.
Article in English | MEDLINE | ID: mdl-34338282

ABSTRACT

Mutations in ITPR1 cause ataxia and aniridia in individuals with Gillespie syndrome (GLSP). However, the pathogenic mechanisms underlying aniridia remain unclear. We identified a de novo GLSP mutation hotspot in the 3'-region of ITPR1 in five individuals with GLSP. Furthermore, RNA-sequencing and immunoblotting revealed an eye-specific transcript of Itpr1, encoding a 218amino acid isoform. This isoform is localized not only in the endoplasmic reticulum, but also in the nuclear and cytoplasmic membranes. Ocular-specific transcription was repressed by SOX9 and induced by MAF in the anterior eye segment (AES) tissues. Mice lacking seven base pairs of the last Itpr1 exon exhibited ataxia and aniridia, in which the iris lymphatic vessels, sphincter and dilator muscles, corneal endothelium and stroma were disrupted, but the neural crest cells persisted after completion of AES formation. Our analyses revealed that the 218-amino acid isoform regulated the directionality of actin fibers and the intensity of focal adhesion. The isoform might control the nuclear entry of transcriptional regulators, such as YAP. It is also possible that ITPR1 regulates both AES differentiation and muscle contraction in the iris.


Subject(s)
Aniridia/blood , Aniridia/genetics , Anterior Eye Segment/growth & development , Cerebellar Ataxia/blood , Cerebellar Ataxia/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Intellectual Disability/blood , Intellectual Disability/genetics , Mutation , Neural Crest/growth & development , Adolescent , Animals , Anterior Eye Segment/metabolism , Child , Child, Preschool , Disease Models, Animal , Exons , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , Neural Crest/metabolism , Protein Isoforms/metabolism , Transfection , Young Adult
9.
Adv Protein Chem Struct Biol ; 127: 271-290, 2021.
Article in English | MEDLINE | ID: mdl-34340770

ABSTRACT

Pseudoexfoliation syndrome (PEX) is characterized by the production of white extracellular fluffy clumps of microfibrillar material that aggregates in various organs throughout the body but is known to cause disease in the eye. The accumulation of PEX material (PEXM) in the anterior segment ocular structures is believed to cause an increase in intraocular pressure (IOP) resulting in pseudoexfoliation glaucoma (PEXG). The onset of PEXG is often bilateral but asymmetric-one eye often presents with glaucoma prior to the other eye. Proteomics has been used to identify key proteins involved in PEXM formation with the end goal of developing effective treatments for PEX and PEXG which may act through inhibiting the formation of the PEX aggregates. To date, a variety of proteins with various molecular functions have been identified from extracted anterior segment structures and fluids, such as aqueous humor (AH) and blood serum of patients affected by PEX. From past studies, some proteins identified in AH, lens capsule epithelium, iris tissue, and blood serum samples include vitamin D binding protein (GC), apolipoprotein A4 (APOA4), lysyl oxidase like-1 (LOXL1), complement C3, beta-crystalline B1, and B2, and antithrombin-III (SERPINC1). Each of these proteins have been observed in eyes with PEX at varying levels within the different eye structures. In this review, we further examine the anterior segment ocular proteomics of PEXM from past studies to better understand the mechanism of PEX and PEXG development. Both genetic and environmental risk factors have been implicated to be involved in the development of PEX and PEXG. This field is at an early stage of investigation identifying how these factors modify proteins both at the expression and functional level to cause changes leading to the pathophysiology of PEX glaucoma.


Subject(s)
Anterior Eye Segment/metabolism , Exfoliation Syndrome/metabolism , Eye Proteins/metabolism , Proteomics , Humans
10.
Invest Ophthalmol Vis Sci ; 62(9): 21, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34259818

ABSTRACT

Purpose: The purpose of this study was to evaluate the role of the canonical Wnt signaling in the development of the myopia. Methods: Plasma from adult patients with myopia, myopic animal models including the adenomatous polyposis coli (APC) gene mutation mouse model, and the form deprivation (FD) induced mouse model of myopia were used. Niclosamide, a canonical Wnt pathway inhibitor, was orally administrated in animal models. Plasma levels of DKK-1 were determined by using enzyme-linked immunosorbent assay. Refraction, vitreous chamber depth (VCD), axial length (AL), and other parameters, were measured at the end of the FD treatment. Canonical Wnt signaling changes were evaluated by Western blot analysis and immunostaining analysis. Results: Plasma level of Wnt inhibitor DKK-1 was markedly decreased in patients with myopia. Meanwhile, the canonical Wnt pathway was progressively activated during myopia development in mice. Moreover, inhibition of canonical Wnt signaling by niclosamide in mouse models markedly reduced lens thickness (LT), VCD, and AL elongation, resulting in myopia inhibition. Conclusions: Dysregulation of canonical Wnt signaling is a characteristic of myopia and targeting Wnt signaling pathways has potential as a therapeutic strategy for myopia.


Subject(s)
Anterior Eye Segment/metabolism , Myopia/genetics , Posterior Eye Segment/metabolism , Refraction, Ocular/physiology , Wnt Signaling Pathway/genetics , Adolescent , Adult , Animals , Anterior Eye Segment/diagnostic imaging , Anterior Eye Segment/drug effects , Biomarkers/metabolism , Disease Models, Animal , Female , Humans , Intercellular Signaling Peptides and Proteins/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Myopia/metabolism , Myopia/physiopathology , Posterior Eye Segment/diagnostic imaging , Posterior Eye Segment/drug effects , Sensory Deprivation , Young Adult
11.
Cornea ; 40(11): 1487-1490, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-33859085

ABSTRACT

PURPOSE: The purpose of this study was to report the clinicopathological features of Peters anomaly in a child with nail-patella syndrome. METHODS: Nail-patella syndrome (NPS) is a rare autosomal dominant connective tissue disorder characterized by several anomalies of the extremities, joints and nails, glomerulopathy, and rarely ocular involvement. NPS is caused by heterozygous loss-of-functional mutations in the LMX1B gene that encodes the LIM homeodomain proteins. RESULTS: This case reports a new association of Peters anomaly in a child with NPS that also had classic skeletal/nail anomalies and protein losing nephropathy. Furthermore, DNA sequence analysis identified a novel missense heterozygous mutation in the LMX1B gene (Transcript ID: NM_001174146) resulting in the replacement of tryptophan by serine in codon 266, suggesting that the mutation (p.Trp.266Ser) affects LMX1B function by disturbing its interactions with other proteins. To the best of our knowledge, this association of Peters anomaly is novel and has not been reported earlier in the ophthalmic and systemic literature on NPS. CONCLUSION: The corneal findings in our case with NPS are similar to those seen in congenital corneal opacification because of Peters anomaly. This novel association of Peters anomaly with NPS may be related to the effects of the LMX1B mutation on corneal development.


Subject(s)
Abnormalities, Multiple , Anterior Eye Segment/abnormalities , Corneal Opacity/genetics , Eye Abnormalities/genetics , LIM-Homeodomain Proteins/genetics , Mutation, Missense , Nail-Patella Syndrome/genetics , Anterior Eye Segment/metabolism , Corneal Opacity/metabolism , Eye Abnormalities/metabolism , Humans , Infant , LIM-Homeodomain Proteins/metabolism , Male , Nail-Patella Syndrome/metabolism , Phenotype
12.
Curr Eye Res ; 46(8): 1075-1088, 2021 08.
Article in English | MEDLINE | ID: mdl-33474991

ABSTRACT

Purpose: To summarize the Integrated Stress Response (ISR) in the context of ophthalmology, with special interest on the cornea and anterior segment. Results: The ISR is a powerful and conserved signaling pathway that allows for cells to respond to a diverse array of both intracellular and extracellular stressors. The pathway is classically responsible for coordination of the cellular response to amino acid starvation, ultraviolet light, heme dysregulation, viral infection, and unfolded protein. Under normal circumstances, it is considered pro-survival and a necessary mechanism through which protein translation is controlled. However, in cases of severe or prolonged stress the pathway can promote apoptosis, and loss of normal cellular phenotype. The activation of this pathway culminates in the global inhibition of cap-dependent protein translation and the canonical expression of the activating transcription factor 4 (ATF4). Conclusion:The eye is uniquely exposed to ISR responsive stressors due to its environmental exposure and relative isolation from the circulatory system which are necessary for its function. We will discuss how this pathway is critical for the proper function of the tissue, its role in development, as well as how targeting of the pathway could alleviate key aspects of diverse ophthalmic diseases.


Subject(s)
Cornea/metabolism , Ophthalmology , Stress, Physiological/physiology , Activating Transcription Factor 4/metabolism , Anterior Eye Segment/metabolism , Humans , Oxidative Stress , Signal Transduction
13.
Biomed Res Int ; 2020: 8206849, 2020.
Article in English | MEDLINE | ID: mdl-33381584

ABSTRACT

The role of the IκB/NF-κB signaling pathway in the uveoscleral outflow pathway was investigated with IκBα gene silencing mediated by the 3-(dimethylamino)-1-propylamine-conjugated glycogen (DMAPA-Glyp) derivative. The IκBα-siRNA-loaded DMAPA-Glyp complex was transfected into the ciliary muscles of rats by intracameral injection (labeled as the DMAPA-Glyp+siRNA group). The Lipofectamine™ 2000 (Lipo)/siRNA complex and the naked siRNA were set as the controls. The mRNA and protein expression of IκBα, NF-κBp65, and MMP-2 were analyzed by real-time PCR, western blotting, and in situ gelatin zymography. Nuclear translocation of NF-κBp65 was analyzed by immunofluorescence. Rat intraocular pressure (IOP) was monitored pre- and postinjection. Gene transfection efficiency and toxicity of the DMAPA-Glyp derivative were also evaluated. After RNA interference (RNAi), IκBα mRNA and protein expression were significantly inhibited. NF-κBp65 mRNA and protein expression showed no significant differences. Nevertheless, nuclear translocation of NF-κBp65 occurred in the DMAPA-Glyp+siRNA group. Both mRNA expression and activity of MMP-2 increased, with the largest increase in the DMAPA-Glyp+siRNA group. IOP in the DMAPA-Glyp+siRNA group fell to the lowest level on day 3 after RNAi. The levels of Cy3-siRNA in the ciliary muscle of the DMAPA-Glyp+siRNA group did not significantly decrease over time. At 7 and 14 d after RNAi, no significant pathological damage was detectable in the eyes injected with the DMAPA-Glyp derivative or the DMAPA-Glyp/siRNA complex. Taken together, our results suggest that downregulation of IκBα expression in the ciliary muscle plays a crucial role in reducing the IOP values of rats. IκBα may become a new molecular target for lowering IOP in glaucoma. The DMAPA-Glyp derivative is safe and feasible as an effective siRNA vector in rat eyes.


Subject(s)
Anterior Eye Segment , Aqueous Humor , Gene Silencing/physiology , I-kappa B Proteins , Signal Transduction/genetics , Animals , Anterior Eye Segment/metabolism , Anterior Eye Segment/physiology , Aqueous Humor/metabolism , Aqueous Humor/physiology , Diamines/chemistry , Drug Delivery Systems , Glycogen/chemistry , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Male , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Transfection
14.
Ophthalmic Physiol Opt ; 40(5): 567-576, 2020 09.
Article in English | MEDLINE | ID: mdl-32839973

ABSTRACT

PURPOSE: To investigate the inhibitory effect of bendazol on form-deprivation myopia (FDM) in rabbits as well as the underlying biochemical processes. METHODS: Forty-eight 3-week-old New Zealand white rabbits were randomly assigned to three groups: a control group, a form-deprivation (FD) group and an FD + bendazol group (treated with 1% bendazol in the FD eyes). Refraction, corneal curvature, vitreous chamber depth (VCD) and axial length (AL) were assessed using streak retinoscopy, keratometry, and A-scan ultrasonography, respectively. Eyeballs were enucleated for histological analysis, and ocular tissues were homogenized to determine the mRNA and protein expression of hypoxia-inducible factor-1α (HIF-1α) and muscarinic acetylcholine receptors (mAChRs). RESULTS: Bendazol inhibited the progression of FDM and suppressed the upregulation of HIF-1α. At week 6, in the control, FD and FD + bendazol groups, the refraction values were 1.38 ± 0.43, 0.03 ± 0.47 and 1.25 ± 0.35 D, respectively (p < 0.001); the ALs were 13.91 ± 0.11, 14.15 ± 0.06 and 13.97 ± 0.10 mm, respectively (p < 0.001) and the VCDs were 6.56 ± 0.06, 6.69 ± 0.07 and 6.61 ± 0.06 mm, respectively (p < 0.001). HIF-1α was upregulated in FD eyes but downregulated in FD + bendazol eyes, while the mAChRs were the opposite. CONCLUSIONS: In the FD rabbit model, bendazol significantly inhibits the development of myopia and downregulates HIF-1α expression, which may provide a novel therapeutic approach for myopia control.


Subject(s)
Anterior Eye Segment , Benzimidazoles , Hypoxia-Inducible Factor 1, alpha Subunit , Myopia, Degenerative , Animals , Rabbits , Anterior Eye Segment/metabolism , Antihypertensive Agents/administration & dosage , Benzimidazoles/administration & dosage , Biomarkers/metabolism , Cornea/metabolism , Cornea/pathology , Disease Models, Animal , Disease Progression , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myopia, Degenerative/diagnosis , Myopia, Degenerative/drug therapy , Myopia, Degenerative/metabolism , Ophthalmic Solutions
15.
Exp Eye Res ; 199: 108200, 2020 10.
Article in English | MEDLINE | ID: mdl-32858007

ABSTRACT

Prion diseases are invariably fatal neurodegenerative disorders that have gained much publicity due to their transmissible nature. Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common human prion disorder, with an incidence of 1 in a million. Inherited prion disorders are relatively rare, and associated with mutations in the prion protein gene. More than 50 different point mutations, deletions, and insertions have been identified so far. Most are autosomal dominant and fully penetrant. Prion disorders also occur in animals, and are of major concern because of the potential for spreading to humans. The principal pathogenic event underlying all prion disorders is a change in the conformation of prion protein (PrPC) from a mainly α-helical to a ß-sheet rich isoform, PrP-scrapie (PrPSc). Accumulation of PrPSc in the brain parenchyma is the major cause of neuronal degeneration. The mechanism by which PrPSc is transmitted, propagates, and causes neurodegenerative changes has been investigated over the years, and several clues have emerged. Efforts are also ongoing for identifying specific and sensitive diagnostic tests for sCJD and animal prion disorders, but success has been limited. The eye is suitable for these evaluations because it shares several anatomical and physiological features with the brain, and can be observed in vivo during disease progression. The retina, considered an extension of the central nervous system, is involved extensively in prion disorders. Accordingly, Optical Coherence Tomography and electroretinogram have shown some promise as pre-mortem diagnostic tests for human and animal prion disorders. However, a complete understanding of the physiology of PrPC and pathobiology of PrPSc in the eye is essential for developing specific and sensitive tests. Below, we summarize recent progress in ocular physiology and pathology in prion disorders, and the eye as an anatomically accessible site to diagnose, monitor disease progression, and test therapeutic options.


Subject(s)
Anterior Eye Segment/metabolism , Gene Expression Regulation , Prions/genetics , Animals , Anterior Eye Segment/pathology , Homeostasis , Humans , Prion Diseases/genetics , Prion Diseases/metabolism , Prion Diseases/pathology , Prions/biosynthesis , Protein Conformation
16.
PLoS Genet ; 16(6): e1008774, 2020 06.
Article in English | MEDLINE | ID: mdl-32555736

ABSTRACT

Cranial neural crest (NC) contributes to the developing vertebrate eye. By multidimensional, quantitative imaging, we traced the origin of the ocular NC cells to two distinct NC populations that differ in the maintenance of sox10 expression, Wnt signalling, origin, route, mode and destination of migration. The first NC population migrates to the proximal and the second NC cell group populates the distal (anterior) part of the eye. By analysing zebrafish pax6a/b compound mutants presenting anterior segment dysgenesis, we demonstrate that Pax6a/b guide the two NC populations to distinct proximodistal locations. We further provide evidence that the lens whose formation is pax6a/b-dependent and lens-derived TGFß signals contribute to the building of the anterior segment. Taken together, our results reveal multiple roles of Pax6a/b in the control of NC cells during development of the anterior segment.


Subject(s)
Anterior Eye Segment/metabolism , Neural Crest/metabolism , Neurogenesis , PAX6 Transcription Factor/metabolism , Zebrafish Proteins/metabolism , Animals , Anterior Eye Segment/cytology , Anterior Eye Segment/embryology , Cell Movement , Mutation , Neural Crest/cytology , Neural Crest/embryology , Neurons/cytology , Neurons/metabolism , PAX6 Transcription Factor/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism , Zebrafish , Zebrafish Proteins/genetics
17.
Exp Eye Res ; 197: 108046, 2020 08.
Article in English | MEDLINE | ID: mdl-32376472

ABSTRACT

Segmental flow in the human trabecular meshwork is a well-documented phenomenon but in depth mechanistic investigations of high flow (HF) and low flow (LF) regions are restricted due to the small amount of tissue available from a single donor. To address this issue we have generated and characterized multiple paired HF and LF cell strains. Here paired HF and LF cell strains were generated from single donors. Cells were characterized for growth and proliferation, as well as gene and protein expression of potential segmental region markers. Cells isolated from HF and LF regions have similar growth and proliferation rates. Gene expression data reveals vascular cell adhesion protein 1 (VCAM1), thrombospondin 2 (THBS2), and tissue inhibitor of metalloproteinase 1 (TIMP1) are potential markers of LF cells in vitro. Protein expression of VCAM1, THBS2 and TIMP1 are complex and may reflect the dynamic nature of the TM. Initial protein expression levels of these genes is either similar between HF and LF cells (VCAM1, THBS2), or higher in HF compared to LF in some strains (TIMP1). However, after long term culture LF cells express higher levels of VCAM1, TIMP1 and THBS2 protein compared to HF cells. HF and LF cell strains are a powerful new tool that enable understanding segmental flow allowing for multiple experiments on the same genetic background.


Subject(s)
Aqueous Humor/metabolism , Glaucoma/diagnosis , Intraocular Pressure/physiology , Trabecular Meshwork/pathology , Aged , Anterior Eye Segment/metabolism , Anterior Eye Segment/pathology , Anterior Eye Segment/physiopathology , Female , Glaucoma/metabolism , Glaucoma/physiopathology , Humans , Male , Microscopy, Confocal , Middle Aged , Trabecular Meshwork/metabolism
18.
Int Ophthalmol ; 40(4): 891-899, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31894458

ABSTRACT

OBJECTIVE: The present study aims to determine hydrogen sulfide (H2S) concentrations of the aqueous humor from patients with diabetic retinopathy (DR) to compare its levels in the anterior segments, also to investigate its effect on the retinal microvascular endothelial cells under high glucose condition. METHODS: AH samples were collected from patients with proliferative diabetic retinopathy (n = 11), non-proliferative diabetic retinopathy (n = 12) and diabetic patients without DR as controls (n = 12). There were 5 patients with PDR received intraocular anti-VEGF injection (Lucentis). Cultured RF/6A cells were grouped into control group, mannitol group, high glucose group and NaHS co-administrated high glucose group. Concentrations of H2S were detected by chemical assay. Cell apoptosis was evaluated by flow cytometry. RESULTS: A significantly higher H2S level was observed in AH samples of PDR patients among other groups. The H2S level of DR group was higher than that of control group. Decreased H2S levels in the AH of post-injected PDR patients were observed compared with their AH samples before the anti-VEGF injection. In cell culture, low concentration of NaHS can reverse high-glucose-induced apoptosis of RF/6A cells. CONCLUSION: Our study revealed increased H2S levels in the anterior segments of different DR patients. The anti-VEGF injection reduced the H2S level in AH from PDR patients. The study suggested that H2S may serve as a biomarker in the progression of PDR. On the other hand, the H2S donor exerted a protective effect on retinal vascular endothelial cells against high-glucose-induced apoptosis.


Subject(s)
Anterior Eye Segment/metabolism , Diabetic Retinopathy/metabolism , Hydrogen Sulfide/metabolism , Aqueous Humor/metabolism , Biomarkers/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
19.
Exp Eye Res ; 190: 107890, 2020 01.
Article in English | MEDLINE | ID: mdl-31811823

ABSTRACT

PURPOSE: The avascular cornea, trabecular meshwork (TM), and lens obtain iron, an essential biometal, from the aqueous humor (AH). The mechanism by which this exchange is regulated, however, is unclear. Recently we reported that non-pigmented ciliary epithelial cells express ferroportin (Fpn) (Ashok, 2018b), an iron export protein modulated by hepcidin, the master regulator of iron homeostasis secreted mainly by the liver. Here, we explored whether ciliary epithelial and other cells in the anterior segment synthesize hepcidin, suggesting local regulation of iron exchange at this site. METHODS: Human and bovine eyes were dissected to isolate the ciliary body (CB), corneal endothelial (CE), TM, lens epithelial (LE), and outer epithelial cell layer of the iris. Total mRNA and protein lysates were processed to evaluate the synthesis and expression of hepcidin, the iron regulatory peptide hormone, Fpn, the only known iron export protein, ceruloplasmin (Cp), a ferroxidase necessary for iron export, transferrin receptor (TfR), a major iron uptake protein, and ferritin, a major iron storage protein. A combination of techniques including reverse transcription polymerase chain reaction (RT-PCR) of total mRNA, Western blotting of protein lysates, and immunofluorescence of fixed tissue sections were used to accomplish these goals. RESULTS: RT-PCR of isolated tissue samples revealed hepcidin-specific mRNA in the CB, TM, CE, and LE of the bovine eye. Western blotting of protein lysates from these tissues showed reactivity for hepcidin, Fpn, ferritin, and TfR. Western blotting and immunohistochemistry of similar tissues isolated from cadaveric human eyes showed expression of hepcidin, Fpn, and Cp in these samples. Notably, Fpn and Cp were expressed on the basolateral membrane of non-pigmented ciliary epithelial cells, facing the AH. CONCLUSIONS: Synthesis and expression of hepcidin and Fpn in the ciliary epithelium suggests local regulation of iron transport from choroidal plexus in the ciliary body to the AH across the blood-aqueous barrier. Expression of hepcidin and Fpn in CE, TM, and LE cells indicates additional regulation of iron exchange between the AH and cornea, TM, and lens, suggesting autonomous regulation of iron homeostasis in the anterior segment. Physiological and pathological implications of these observations are discussed.


Subject(s)
Anterior Eye Segment/metabolism , Anti-Infective Agents/metabolism , Hepcidins/biosynthesis , Adult , Aged , Animals , Blotting, Western , Cation Transport Proteins/metabolism , Cattle , Ceruloplasmin/metabolism , Ciliary Body/metabolism , Electrophoresis, Polyacrylamide Gel , Endothelium, Corneal/metabolism , Epithelial Cells/metabolism , Female , Fluorescent Antibody Technique, Indirect , Hepcidins/genetics , Humans , Iris/metabolism , Lens, Crystalline/metabolism , Male , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trabecular Meshwork/metabolism
20.
Exp Eye Res ; 190: 107888, 2020 01.
Article in English | MEDLINE | ID: mdl-31786158

ABSTRACT

Elevated intraocular pressure (IOP) is the primary risk factor for glaucoma and is the only treatable feature of the disease. There is a correlation between elevated pressure and homeostatic reductions in the aqueous humor outflow resistance via changes in the extracellular matrix of the trabecular meshwork. It is unclear how these extracellular matrix changes affect segmental patterns of aqueous humor outflow, nor do we understand their causal relationship. The goal of this study was to determine whether there are changes in the segmental outflow regions with perfusion in normal eyes, and whether these regions change during the IOP homeostatic response to elevated pressure. Using human anterior segment perfusion organ culture, we measured the amount of high flow (HF), intermediate flow (MF), and low flow (LF) regions before and after 7 days of perfusion at either physiologic pressure ("1x") or at elevated pressure ("2x"). We found a small but significant decrease in the amount of HF regions over 7 days perfusion at 1x pressure, and a twofold increase in the amount of MF regions over 7 days perfusion at 2x pressure. Small positional differences, or shifts in the specific location of HF, MF, or LF, occurred on a per eye basis and were not found to be statistically significant across biological replicates. Differences in the amount of segmental flow regions of contralateral eyes flowed at 1x pressure for 7 days were small and not statistically significant. These results demonstrate that perfusion at physiologic pressure had little effect on the distribution and amount of HF, MF and LF regions. However, the overall amount of MF regions is significantly increased in response to perfusion at elevated pressure during IOP homeostatic resistance adjustment. The amount of both HF and LF regions was decreased accordingly suggesting a coordinated response in the TM to elevated pressure.


Subject(s)
Anterior Eye Segment/metabolism , Aqueous Humor/physiology , Intraocular Pressure/physiology , Ocular Hypertension/metabolism , Trabecular Meshwork/metabolism , Aged , Aged, 80 and over , Female , Humans , Male , Organ Culture Techniques , Tissue Donors
SELECTION OF CITATIONS
SEARCH DETAIL