Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Biomolecules ; 13(12)2023 12 09.
Article in English | MEDLINE | ID: mdl-38136638

ABSTRACT

Fibrotic cataracts, posterior capsular opacification (PCO), and anterior subcapsular cataracts (ASC) are mainly attributed to the transforming growth factor-ß (TGFß)-induced epithelial-to-mesenchymal transition (EMT) of lens epithelial cells (LECs). Previous investigations from our laboratory have shown the novel role of non-canonical TGFß signaling in the progression of EMT in LECs. In this study, we have identified YAP as a critical signaling molecule involved in lens fibrosis. The observed increase in nuclear YAP in capsules of human ASC patients points toward the involvement of YAP in lens fibrosis. In addition, the immunohistochemical (IHC) analyses on ocular sections from mice that overexpress TGFß in the lens (TGFßtg) showed a co-expression of YAP and α-SMA in the fibrotic plaques when compared to wild-type littermate lenses, which do not. The incubation of rat lens explants with verteporfin, a YAP inhibitor, prevented a TGFß-induced fiber-like phenotype, α-SMA, and fibronectin expression, as well as delocalization of E-cadherin and ß-catenin. Finally, LECs co-incubated with TGFß and YAP inhibitor did not exhibit an induction in matrix metalloproteinase 2 compared to those LECs treated with TGFß alone. In conclusion, these data demonstrate that YAP is required for TGFß-mediated lens EMT and fibrosis.


Subject(s)
Capsule Opacification , Lens, Crystalline , Humans , Rats , Animals , Mice , Matrix Metalloproteinase 2/metabolism , YAP-Signaling Proteins , Lens, Crystalline/metabolism , Epithelial Cells/metabolism , Capsule Opacification/pathology , Transforming Growth Factor beta/metabolism , Fibrosis
2.
Front Cell Dev Biol ; 10: 886053, 2022.
Article in English | MEDLINE | ID: mdl-35656546

ABSTRACT

Injury to the ocular lens perturbs cell-cell and cell-capsule/basement membrane interactions leading to a myriad of interconnected signaling events. These events include cell-adhesion and growth factor-mediated signaling pathways that can ultimately result in the induction and progression of epithelial-mesenchymal transition (EMT) of lens epithelial cells and fibrosis. Since the lens is avascular, consisting of a single layer of epithelial cells on its anterior surface and encased in a matrix rich capsule, it is one of the most simple and desired systems to investigate injury-induced signaling pathways that contribute to EMT and fibrosis. In this review, we will discuss the role of key cell-adhesion and mechanotransduction related signaling pathways that regulate EMT and fibrosis in the lens.

3.
J Neurosci Res ; 100(2): 638-652, 2022 02.
Article in English | MEDLINE | ID: mdl-34822722

ABSTRACT

Glaucoma is one of the leading causes of irreversible blindness and can result from abnormalities in anterior segment structures required for aqueous humor outflow, including the trabecular meshwork (TM) and Schlemm's canal (SC). Transcription factors such as AP-2ß play critical roles in anterior segment development. Here, we show that the Mgp-Cre knock-in (Mgp-Cre.KI) mouse can be used to target the embryonic periocular mesenchyme giving rise to the TM and SC. Fate mapping of male and female mice indicates that AP-2ß loss causes a decrease in iridocorneal angle cells derived from Mgp-Cre.KI-expressing populations compared to controls. Moreover, histological analyses revealed peripheral iridocorneal adhesions in AP-2ß mutants that were accompanied by a decrease in expression of TM and SC markers, as observed using immunohistochemistry. In addition, rebound tonometry showed significantly higher intraocular pressure (IOP) that was correlated with a progressive significant loss of retinal ganglion cells, reduced retinal thickness, and reduced retinal function, as measured using an electroretinogram, in AP-2ß mutants compared with controls, reflecting pathology described in late-stage glaucoma patients. Importantly, elevated IOP in AP-2ß mutants was significantly reduced by treatment with latanoprost, a prostaglandin analog that increases unconventional outflow. These findings demonstrate that AP-2ß is critical for TM and SC development, and that these mutant mice can serve as a model for understanding and treating progressive human primary angle-closure glaucoma.


Subject(s)
Glaucoma , Trabecular Meshwork , Transcription Factor AP-2 , Animals , Aqueous Humor/metabolism , Female , Glaucoma/genetics , Glaucoma/metabolism , Humans , Intraocular Pressure , Male , Mice , Trabecular Meshwork/metabolism , Trabecular Meshwork/pathology , Transcription Factor AP-2/genetics
4.
Int J Mol Sci ; 22(21)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34769418

ABSTRACT

Fibrotic cataracts have been attributed to transforming growth factor-beta (TGF-ß)-induced epithelial-to-mesenchymal transition (EMT). Using mouse knockout (KO) models, our laboratory has identified MMP9 as a crucial protein in the TGF-ß-induced EMT process. In this study, we further revealed an absence of alpha-smooth muscle actin (αSMA) and filamentous-actin (F-actin) stress fibers in MMP9KO mouse lens epithelial cell explants (LECs). Expression analysis using NanoString revealed no marked differences in αSMA (ACTA2) and beta-actin (ß-actin) (ACTB) mRNA between the lenses of TGF-ß-overexpressing (TGF-ßtg) mice and TGF-ßtg mice on a MMP9KO background. We subsequently conducted a protein array that revealed differential regulation of proteins known to be involved in actin polymerization and cell migration in TGF-ß-treated MMP9KO mouse LECs when compared to untreated controls. Immunofluorescence analyses using rat LECs and the novel MMP9-specific inhibitor, JNJ0966, revealed similar differential regulation of cortactin, FAK, LIMK1 and MLC2 as observed in the array. Finally, a reduction in the nuclear localization of MRTF-A, a master regulator of cytoskeletal remodeling during EMT, was observed in rat LECs co-treated with JNJ0966 and TGF-ß. In conclusion, MMP9 deficiency results in differential regulation of proteins involved in actin polymerization and cell migration, and this in turn prevents TGF-ß-induced EMT in the lens.


Subject(s)
Actins/metabolism , Lens, Crystalline/metabolism , Matrix Metalloproteinase 9/metabolism , Proteome/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Movement/physiology , Disease Models, Animal , Epithelial-Mesenchymal Transition , Lens, Crystalline/pathology , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Mice, Transgenic , Polymerization , Transcriptome
5.
Int J Mol Sci ; 22(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34445433

ABSTRACT

The cornea is an anterior eye structure specialized for vision. The corneal endothelium and stroma are derived from the periocular mesenchyme (POM), which originates from neural crest cells (NCCs), while the stratified corneal epithelium develops from the surface ectoderm. Activating protein-2ß (AP-2ß) is highly expressed in the POM and important for anterior segment development. Using a mouse model in which AP-2ß is conditionally deleted in the NCCs (AP-2ß NCC KO), we investigated resulting corneal epithelial abnormalities. Through PAS and IHC staining, we observed structural and phenotypic changes to the epithelium associated with AP-2ß deletion. In addition to failure of the mutant epithelium to stratify, we also observed that Keratin-12, a marker of the differentiated epithelium, was absent, and Keratin-15, a limbal and conjunctival marker, was expanded across the central epithelium. Transcription factors PAX6 and P63 were not observed to be differentially expressed between WT and mutant. However, growth factor BMP4 was suppressed in the mutant epithelium. Given the non-NCC origin of the epithelium, we hypothesize that the abnormalities in the AP-2ß NCC KO mouse result from changes to regulatory signaling from the POM-derived stroma. Our findings suggest that stromal pathways such as Wnt/ß-Catenin signaling may regulate BMP4 expression, which influences cell fate and stratification.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Down-Regulation , Epithelium, Corneal/abnormalities , Gene Deletion , Transcription Factor AP-2/genetics , Animals , Bone Morphogenetic Protein 4/genetics , Cell Differentiation , Epithelium, Corneal/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Keratin-12/metabolism , Keratin-15/metabolism , Male , Mice , Neural Crest/metabolism , Phenotype , Transcription Factor AP-2/metabolism , Wnt Signaling Pathway
6.
Curr Eye Res ; 46(10): 1509-1515, 2021 10.
Article in English | MEDLINE | ID: mdl-33689532

ABSTRACT

Purpose: Our lab has shown that conditionally disrupting the transcription factor activating protein 2ß (Tfap2b) gene, responsible for the activating protein-2ß (AP-2ß) transcription factor, exclusively in cranial neural crest cells (AP-2ß NCC KO), leads to anterior segment dysgenesis and a closed angle phenotype. The purpose of the current study is to determine if there is a progressive loss of retinal ganglion cells (RGCs) in the mutant over time and whether this loss was associated with macroglial activity changes and elevated intraocular pressure (IOP).Methods: Using the Cre-loxP system, we generated a conditional knockout of Tfap2b exclusively in cranial NCC (AP-2ß NCC KO). Immunohistochemistry was performed using anti-Brn3a, anti-GFAP and anti-Vimentin antibodies. IOP was measured using a tonometer and the data was analyzed using GraphPad Prism software. Brn3a and DAPI positive cells were counted using Image-J and statistical analysis was performed with GraphPad Prism software.Results: Our findings revealed that while no statistical difference in Brn3a expression was observed between wild-type and mutant mice at postnatal day (P) 4 or P10, at P40 (p < .01) and P42 (p < .0001) Brn3a expression was significantly reduced in the mutant retina at the region of the ONH. There was also increased expression of glial fibrillary acidic protein (GFAP) by Müller cells in the AP-2ß NCC KO mice at P35 and P40, indicating the presence of neuroinflammation. Moreover, increased IOP was observed starting at P35 and continuing at P40 and P42 (p < .0001 for all three ages examined).Conclusions: Together, these findings suggest that the retinal damage observed in the KO mouse becomes apparent by P40 after increased IOP was observed at P35 and progressed over time. The AP-2ß NCC KO mouse may therefore be a novel experimental model for glaucoma.


Subject(s)
Glaucoma/diagnosis , Neural Crest/metabolism , Retinal Diseases/diagnosis , Retinal Ganglion Cells/pathology , Transcription Factor AP-2/genetics , Animals , Disease Progression , Electrophoresis , Glaucoma/genetics , Glaucoma/metabolism , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Intraocular Pressure/physiology , Mice , Mice, Knockout , Microglia/pathology , Polymerase Chain Reaction , Retinal Diseases/genetics , Retinal Diseases/metabolism , Tonometry, Ocular , Transcription Factor Brn-3A/metabolism , Vimentin/metabolism
7.
Exp Eye Res ; 201: 108231, 2020 12.
Article in English | MEDLINE | ID: mdl-33039457

ABSTRACT

The cornea is a highly specialized transparent tissue located at the anterior most surface of the eye. It consists of three main layers, the outer stratified squamous epithelium, the inner endothelium, and the intermediate stroma. Formation of these layers during development involves a complex interaction between ectodermal-derived structures, such as the overlying head ectoderm with the periocular mesenchyme (POM), the latter of which is comprised of neural crest cells (NCC) and mesoderm-derived progenitor cells. Regulation of corneal epithelial development, including both epithelial cell fate and stratification, has been shown to depend on numerous bi-directional mesenchymal-epithelial signaling pathways. In this review we pay particular attention to the genes and signaling pathways that involve the POM.


Subject(s)
Cornea/diagnostic imaging , Neural Crest/growth & development , Animals , Cell Differentiation , Cornea/metabolism , Humans , Neural Crest/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
8.
Exp Eye Res ; 195: 108042, 2020 06.
Article in English | MEDLINE | ID: mdl-32353428

ABSTRACT

Previously, we have shown that Tfap2b, the gene encoding transcription factor AP-2ß, is needed for normal mouse eye development. Specifically, targeted loss of Tfap2b in neural crest cells (NCCs)1 and their derivatives, particularly the periocular mesenchyme (POM), resulted in anterior segment defects affecting the cornea and angle tissue. These defects were further associated with an increase in intraocular pressure (IOP). The present study investigates the underlying changes in embryonic and postnatal POM cell development and differentiation caused by loss of AP-2ß in the NCCs, particularly in the structures that control aqueous outflow, using Wnt1Cre+/-; Tfap2b-/lox; tdTomatolox/+ mice (AP-2ß neural crest cell knockout or AP-2ß NCC KO). Toluidine blue-stained sections and ultrathin sections stained with uranyl acetate and lead citrate were used to assess morphology and ultrastructure, respectively. Immunohistochemistry of KO and control eyes was performed at embryonic day (E) 15.5, E18.5, postnatal day (P) 1, P7 and P14 using phospho-histone H3 (PH3), α-smooth muscle actin (α-SMA), myocilin and endomucin antibodies, as well as a TUNEL assay. Conditional deletion of AP-2ß in the NCC-derived POM resulted in defects that appeared during both embryogenesis and postnatal stages. Fate mapping of the knockout cells in the mutants revealed that the POM migrated appropriately into the eye during embryogenesis. However, during postnatal stages a significant reduction in POM proliferation in the angle region was observed in the mutants compared to controls. This was accompanied by a lack of expression of appropriate trabecular meshwork and Schlemm's canal markers. This is the first study to show that AP-2ß is required for development and differentiation of the trabecular meshwork and Schlemm's canal. Together, these defects likely contributed to the elevated intraocular pressure (IOP) previously reported in the AP-2ß NCC KO mice.


Subject(s)
Gene Expression Regulation, Developmental , Intraocular Pressure/physiology , RNA/genetics , Trabecular Meshwork/growth & development , Transcription Factor AP-2/genetics , Animals , Cells, Cultured , Immunohistochemistry , Mice , Mice, Knockout , Models, Animal , RNA/metabolism , Trabecular Meshwork/metabolism , Transcription Factor AP-2/metabolism
9.
Int J Mol Sci ; 20(9)2019 Apr 27.
Article in English | MEDLINE | ID: mdl-31035577

ABSTRACT

Cataracts are the leading cause of blindness worldwide. Although surgery is a successful method to restore vision loss due to cataracts, post-surgical complications can occur, such as secondary cataracts, also known as posterior capsular opacification (PCO). PCO arises when lens epithelial cells (LEC) are left behind in the capsular bag following surgery and are induced to undergo epithelial to mesenchymal transition (EMT). Following EMT, LEC morphology and phenotype are altered leading to a loss of transparency and vision. Transforming growth factor (TGF)-ß-induced signaling through both canonical, TGF-ß/Smad, and non-canonical, ß-catenin/Wnt and Rho/ROCK/MRTF-A, pathways have been shown to be involved in lens EMT, and thus PCO. However, the interactions between these signaling pathways in the lens have not been thoroughly explored. In the current study we use rat LEC explants as an ex vivo model, to examine the interplay between three TGF-ß-mediated pathways using α-smooth muscle actin (α-SMA) as a molecular marker for EMT. We show that Smad3 inhibition via SIS3 prevents nuclear translocation of ß-catenin and MRTF-A, and α-SMA expression, suggesting a key role of Smad3 in regulation of MRTF-A and ß-catenin nuclear transport in LECs. Further, we demonstrate that inhibition of ß-catenin/CBP interaction by ICG-001 decreased the amount of phosphorylated Smad3 upon TGF-ß stimulation in addition to significantly decreasing the expression levels of TGF-ß receptors, TBRII and TBRI. Overall, our findings demonstrate interdependence between the canonical and non-canonical TGF-ß-mediated signaling pathways controlling EMT in the lens.


Subject(s)
Epithelial-Mesenchymal Transition , Lens, Crystalline/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , beta Catenin/metabolism , Animals , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Protein Binding , Protein Transport , Rats , Signal Transduction/drug effects , Smad3 Protein/genetics , Transforming Growth Factor beta/pharmacology , beta Catenin/genetics
10.
J Neurosci Res ; 97(1): 7-15, 2019 01.
Article in English | MEDLINE | ID: mdl-29660784

ABSTRACT

Development of the eye is closely associated with neural crest cell migration and specification. Eye development is extremely complex, as it requires the working of a combination of local factors, receptors, inductors, and signaling interactions between tissues such as the optic cup and periocular mesenchyme (POM). The POM is comprised of neural crest-derived mesenchymal progenitor cells that give rise to numerous important ocular structures including those tissues that form the optic cup and anterior segment of the eye. A number of genes are involved in the migration and specification of the POM such as PITX2, PITX3, FOXC1, FOXE3, PAX6, LMX1B, GPR48, TFAP2A, and TFAP2B. In this review, we will discuss the relevance of these genes in the development of the POM and how mutations and defects result in rare ocular diseases.


Subject(s)
Eye Abnormalities/genetics , Eye Diseases/genetics , Neural Crest/abnormalities , Neural Crest/metabolism , Rare Diseases/genetics , Anterior Eye Segment/abnormalities , Eye Diseases/pathology , Humans , Mutation , Posterior Eye Segment/abnormalities , Rare Diseases/pathology , Transcription Factors
11.
Invest Ophthalmol Vis Sci ; 59(6): 2229-2239, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29715367

ABSTRACT

Purpose: The combined action of the activating protein-2 (AP-2) transcription factors, AP-2α and AP-2ß, is important in early retinal development, specifically in the formation of horizontal cells. However, in previous studies, it was not possible to analyze postnatal development and function of additional retinal subtypes. Methods: We used a double conditional deletion of AP-2α and AP-2ß from the retina to further examine the combinatory role of these genes in retinal cell patterning and function in postnatal adult mice as measured by Voronoi domain area and nearest-neighbor distance spatial analyses and ERGs, respectively. Results: Conditional deletion of both AP-2α and AP-2ß from the retina resulted in a variety of abnormalities, including the absence of horizontal cells, defects in the photoreceptor ribbons in which synapses failed to form, along with evidence of aberrant amacrine cell arrangement. Although no significant changes in amacrine cell population numbers were observed in the double mutants, significant irregularities in the mosaic patterning of amacrine cells was observed as demonstrated by both Voronoi domain areas and nearest-neighbor distances analyses. These changes were further accompanied by an alteration in the retinal response to light as recorded by ERGs. In particular, in the double-mutant mice lacking AP-2α and AP-2ß, the b-wave amplitude, representative of interneuron signal processing, was significantly reduced compared with control littermates. Conclusions: Together these findings demonstrate the requirement for both AP-2α and AP-2ß in proper amacrine mosaic patterning and a normal functional light response in the retina.


Subject(s)
Amacrine Cells/metabolism , Animals, Newborn , DNA/genetics , Gene Expression Regulation, Developmental , Retina/metabolism , Sequence Deletion , Transcription Factor AP-2/genetics , Amacrine Cells/ultrastructure , Animals , Base Sequence , Cell Count , Electroretinography , Female , Fluorescent Antibody Technique, Indirect , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Models, Animal , Retina/ultrastructure , Transcription Factor AP-2/biosynthesis
12.
Mol Biol Cell ; 28(7): 907-921, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28209733

ABSTRACT

The most common vision-disrupting complication of cataract surgery is posterior capsule opacification (PCO; secondary cataract). PCO is caused by residual lens cells undergoing one of two very different cell fates: either transdifferentiating into myofibroblasts or maturing into lens fiber cells. Although TGFß has been strongly implicated in lens cell fibrosis, the factors responsible for the latter process have not been identified. We show here for the first time that TGFß can induce purified primary lens epithelial cells within the same culture to undergo differentiation into either lens fiber cells or myofibroblasts. Marker analysis confirmed that the two cell phenotypes were mutually exclusive. Blocking the p38 kinase pathway, either with direct inhibitors of the p38 MAP kinase or a small-molecule therapeutic that also inhibits the activation of p38, prevented TGFß from inducing epithelial-myofibroblast transition and cell migration but did not prevent fiber cell differentiation. Rapamycin had the converse effect, linking MTOR signaling to induction of fiber cell differentiation by TGFß. In addition to providing novel potential therapeutic strategies for PCO, our findings extend the so-called TGFß paradox, in which TGFß can induce two disparate cell fates, to a new epithelial disease state.


Subject(s)
Lens, Crystalline/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chick Embryo , Epithelial Cells/metabolism , Epithelium/metabolism , Eye Proteins/metabolism , Humans , Myofibroblasts/metabolism , Signal Transduction
13.
Nephrol Dial Transplant ; 32(1): 50-61, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27190383

ABSTRACT

Background: For patients using peritoneal dialysis (PD), the peritoneal membrane can develop fibrosis and angiogenesis, leading to ultrafiltration failure, chronic hypervolemia and increased risk of technique failure and mortality. Matrix metalloproteinases (MMPs), and specifically the gelatinases (MMP2 and MMP9), may be involved in peritoneal membrane injury. Methods: From stable PD patients, mesothelial cells were assayed for MMP gene expression. MMP9 was overexpressed in mouse peritoneum by adenovirus, and MMP9 -/- mice were subjected to transforming growth factor ß (TGF-ß)-induced peritoneal fibrosis. Results: MMP9 mRNA expression correlated with peritoneal membrane solute transport properties. Overexpression of MMP9 in the mouse peritoneum induced submesothelial thickening and angiogenesis. MMP9 induced mesothelial cell transition to a myofibroblast phenotype measured by increased alpha smooth muscle actin and decreased E-cadherin expression. Angiogenesis was markedly reduced in MMP9 -/- mice treated with an adenovirus expressing active TGF-ß compared with wild-type mice. TGF-ß-mediated E-cadherin cleavage was MMP9 dependent, and E-cadherin cleavage led to ß-catenin-mediated signaling. A ß-catenin inhibitor blocked the angiogenic response induced by AdMMP9. Conclusions: Our data suggest that MMP9 is involved in peritoneal membrane injury possibly through cleavage of E-cadherin and induction of ß-catenin signaling. MMP9 is a potential biomarker for peritoneal membrane injury and is a therapeutic target to protect the peritoneal membrane in PD patients.


Subject(s)
Cadherins/metabolism , Hemodialysis Solutions/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/etiology , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/etiology , beta Catenin/metabolism , Animals , Biological Transport , Cadherins/genetics , Humans , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Signal Transduction/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , beta Catenin/genetics
14.
Invest Ophthalmol Vis Sci ; 57(14): 5984-5995, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27820954

ABSTRACT

PURPOSE: Homeostatic turnover of the trabecular meshwork extracellular matrix (ECM) is essential to regulate aqueous humor outflow and to maintain intraocular pressure homeostasis. In this study, we evaluated aqueous humor turnover, intraocular pressure, and trabecular meshwork organization in MMP-9 null mice. METHODS: Intraocular pressure and aqueous humor turnover were measured in MMP-9 null versus wild-type mice. Morphology of the anterior segment of the eye, with special attention to the structural organization of the trabecular meshwork, was investigated by means of optical coherence tomography, light microscopy, and transmission electron microscopy. Furthermore, using quantitative real-time polymerase chain reaction and immunostainings, we evaluated the ECM composition of the trabecular meshwork. Finally, the integrity and function of the retina and optic nerve were assessed, via optical coherence tomography, histologic techniques, and optomotor testing. RESULTS: MMP-9 null mice displayed early-onset ocular hypertension and reduced aqueous humor turnover. While transmission electron microscopic analysis did not reveal any abnormalities in the cellular organization of the trabecular meshwork, detailed investigation of collagen expression indicated that there is an aberrant trabecular meshwork ECM composition in MMP-9 null mice. Notably, at the age of 13 months, no glaucomatous neurodegeneration was seen in MMP-9 null mice. CONCLUSIONS: Our observations corroborate MMP-9 as an important remodeler of the collagenous composition of the trabecular meshwork and provide evidence for a causal link between MMP-9 deficiency, trabecular meshwork ultrastructure, and ocular hypertension.


Subject(s)
Aqueous Humor/metabolism , Intraocular Pressure/physiology , Matrix Metalloproteinase 9/metabolism , Ocular Hypertension/metabolism , Trabecular Meshwork/metabolism , Animals , Disease Models, Animal , Extracellular Matrix/metabolism , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Ocular Hypertension/physiopathology , Optic Nerve/pathology , Real-Time Polymerase Chain Reaction , Retina/pathology , Tomography, Optical Coherence
15.
Invest Ophthalmol Vis Sci ; 57(13): 5736-5747, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27787561

ABSTRACT

PURPOSE: Transforming growth factor-ß-induced epithelial-mesenchymal transition (EMT) is one of the main causes of posterior capsular opacification (PCO) or secondary cataract; however, the signaling events involved in TGF-ß-induced PCO have not been fully characterized. Here, we focus on examining the role of ß-catenin/cyclic AMP response element-binding protein (CREB)-binding protein (CBP) and ß-catenin/T-cell factor (TCF)-dependent signaling in regulating cytoskeletal dynamics during TGF-ß-induced EMT in lens epithelial explants. METHODS: Rat lens epithelial explants were cultured in medium M199 in the absence of serum. Explants were treated with TGF-ß2 in the presence or absence of the ß-catenin/CBP interaction inhibitor, ICG-001, or the ß-catenin/TCF interaction inhibitor, PNU-74654. Western blot and immunofluorescence experiments were carried out and analyzed. RESULTS: An increase in the expression of fascin, an actin-bundling protein, was observed in the lens explants upon stimulation with TGF-ß, and colocalized with F-actin filaments. Inhibition of ß-catenin/CBP interactions, but not ß-catenin/TCF interactions, led to a decrease in TGF-ß-induced fascin and stress fiber formation, as well as a decrease in the expression of known markers of EMT, α-smooth muscle actin (α-SMA) and matrix metalloproteinase 9 (MMP9). In addition, inhibition of ß-catenin/CBP-dependent signaling also prevented TGF-ß-induced downregulation of epithelial cadherin (E-cadherin) in lens explants. CONCLUSIONS: We show that ß-catenin/CBP-dependent signaling regulates fascin, MMP9, and α-SMA expression during TGF-ß-induced EMT. We demonstrate that ß-catenin/CBP-dependent signaling is crucial for TGF-ß-induced EMT in the lens.


Subject(s)
Capsule Opacification/metabolism , Epithelial-Mesenchymal Transition/drug effects , Transforming Growth Factor beta2/pharmacology , beta Catenin/pharmacology , Actins , Animals , Blotting, Western , Capsule Opacification/pathology , Carrier Proteins/biosynthesis , Carrier Proteins/drug effects , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Microfilament Proteins/biosynthesis , Microfilament Proteins/drug effects , Rats , Rats, Wistar , Recombinant Proteins , Signal Transduction
16.
Mol Med ; 22: 713-723, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27704140

ABSTRACT

Transforming growth factor (TGF)-ß-induced epithelial-mesenchymal transition (EMT) leads to the formation of ocular fibrotic pathologies, such as anterior subcapsular cataract and posterior capsule opacification. Remodeling of the actin cytoskeleton, mediated by the Rho family of GTPases, plays a key role in EMT, however, how actin dynamics affect downstream markers of EMT has not been fully determined. Our previous work suggests that myocardin related transcription factor A (MRTF-A), an actin-binding protein, might be an important mediator of TGFß-induced EMT in lens epithelial cells. The aim of the current study was to determine the requirement of RhoA/ROCK signaling in mediating TGFß-induced nuclear accumulation of MRTF-A, and ultimate expression of α-smooth muscle actin (αSMA), a marker of a contractile, myofibroblast phenotype. Using rat lens epithelial explants, we demonstrate that ROCK inhibition using Y-27632 prevents TGFß-induced nuclear accumulation of MRTF-A, E-cadherin/ß-catenin complex disassembly, and αSMA expression. Using a novel inhibitor specifically targeting MRTF-A signaling, CCG-203971, we further demonstrate the requirement of MRTF-A nuclear localization and activity in the induction of αSMA expression. Overall, our findings suggest that TGFß-induced cytoskeletal reorganization through RhoA/ROCK/MRTF-A signaling is critical to EMT of lens epithelial cells.

17.
Dis Model Mech ; 9(8): 849-61, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27483349

ABSTRACT

Anterior segment dysgenesis (ASD) encompasses a group of developmental disorders in which a closed angle phenotype in the anterior chamber of the eye can occur and 50% of patients develop glaucoma. Many ASDs are thought to involve an inappropriate patterning and migration of the periocular mesenchyme (POM), which is derived from cranial neural crest cells (NCCs) and mesoderm. Although, the mechanism of this disruption is not well understood, a number of transcriptional regulatory molecules have previously been implicated in ASDs. Here, we investigate the function of the transcription factor AP-2ß, encoded by Tfap2b, which is expressed in NCCs and their derivatives. Wnt1-Cre-mediated conditional deletion of Tfap2b in NCCs resulted in post-natal ocular defects typified by opacity. Histological data revealed that the conditional AP-2ß NCC knockout (KO) mutants exhibited dysgenesis of multiple structures in the anterior segment of the eye including defects in the corneal endothelium, corneal stroma, ciliary body and disruption in the iridocorneal angle with adherence of the iris to the cornea. We further show that this phenotype leads to a significant increase in intraocular pressure and a subsequent loss of retinal ganglion cells and optic nerve degeneration, features indicative of glaucoma. Overall, our findings demonstrate that AP-2ß is required in the POM for normal development of the anterior segment of the eye and that the AP-2ß NCC KO mice might serve as a new and exciting model of ASD and glaucoma that is fully penetrant and with early post-natal onset.


Subject(s)
Anterior Eye Segment/abnormalities , Eye Abnormalities/pathology , Gene Deletion , Glaucoma/pathology , Neural Crest/metabolism , Skull/pathology , Transcription Factor AP-2/genetics , Animals , Anterior Eye Segment/embryology , Anterior Eye Segment/pathology , Anterior Eye Segment/physiopathology , Axons/pathology , Cell Count , Cornea/abnormalities , Cornea/embryology , Cornea/pathology , Cornea/physiopathology , Eye Abnormalities/complications , Eye Abnormalities/physiopathology , Glaucoma/complications , Glaucoma/physiopathology , Intraocular Pressure , Mice , Mice, Knockout , Mutation/genetics , Neuroglia/pathology , Optic Nerve/pathology , Retinal Ganglion Cells/metabolism
18.
Invest Ophthalmol Vis Sci ; 57(3): 1072-81, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26968737

ABSTRACT

PURPOSE: The homeodomain transcription factor, PITX2, is at the apex of a genetic pathway required for corneal development, but the critical effector genes regulated by the PITX2 remain unknown. The purpose of this study was to discover and validate PITX2-dependent mechanisms required for specifying cell lineages and establishing angiogenic privilege within the developing cornea. METHODS: Microarrays were used to compare gene expression in corneas isolated from temporal Pitx2 knockout embryos and control littermates. Quantitative RT-PCR and immunohistochemistry was used to further validate Tfap2b expression differences in Pitx2 knockout versus control corneas. In situ hybridization and protein immunohistochemistry were used to assay eyes of a Tfap2b allelic series of embryos to identify differentiated cellular lineages in the cornea, blood vessel endothelium, or lymphatic vessel endothelium. RESULTS: We show that PITX2 is required for the expression of Tfap2b, encoding the AP-2ß transcription factor, in the neural crest during corneal development. Markers of differentiated corneal epithelium and stroma are expressed in the absence of AP-2ß. In contrast, markers of differentiated corneal endothelium are not expressed in the absence of AP-2ß. Endomucin+ blood vessels are present throughout the developing corneal stroma in the absence of AP-2ß, whereas LYVE1+ lymphatic vessels are not found. CONCLUSIONS: The AP-2ß transcription factor is an important effector of PITX2 function during corneal development, required for differentiation of corneal endothelium and establishment of angiogenic privilege. Unlike PITX2, AP-2ß is not required for the early expression of available lineage specific markers for the corneal epithelium and stroma during embryogenesis, nor establishment of lymphangiogenic privilege. Therefore, additional PITX2-dependent factors likely regulate these latter processes during embryonic development. These results extend our understanding of the genetic mechanisms regulating cornea development.


Subject(s)
Endothelium, Corneal/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Morphogenesis/genetics , Neovascularization, Physiologic/genetics , Pregnancy, Animal , Transcription Factor AP-2/genetics , Transcription Factors/genetics , Animals , Cell Differentiation , Cornea/embryology , Cornea/metabolism , Endothelium, Corneal/metabolism , Female , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Pregnancy , Transcription Factor AP-2/metabolism , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Homeobox Protein PITX2
19.
Am J Pathol ; 184(7): 2001-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24814605

ABSTRACT

Epithelial-mesenchymal transition (EMT) is associated with fibrotic diseases in the lens, such as anterior subcapsular cataract (ASC) formation. Often mediated by transforming growth factor (TGF)-ß, EMT in the lens involves the transformation of lens epithelial cells into a multilayering of myofibroblasts, which manifest as plaques beneath the lens capsule. TGF-ß-induced EMT and ASC have been associated with the up-regulation of two matrix metalloproteinases (MMPs): MMP-2 and MMP-9. The current study used MMP-2 and MMP-9 knockout (KO) mice to further determine their unique roles in TGF-ß-induced ASC formation. Adenoviral injection of active TGF-ß1 into the anterior chamber of all wild-type and MMP-2 KO mice led to the formation of distinct ASC plaques that were positive for α-smooth muscle actin, a marker of EMT. In contrast, only a small proportion of the MMP-9 KO eyes injected with adenovirus-expressing TGF-ß1 exhibited ASC plaques. Isolated lens epithelial explants from wild-type and MMP-2 KO mice that were treated with TGF-ß exhibited features indicative of EMT, whereas those from MMP-9 KO mice did not acquire a mesenchymal phenotype. MMP-9 KO mice were further bred onto a TGF-ß1 transgenic mouse line that exhibits severe ASC formation, but shows a resistance to ASC formation in the absence of MMP-9. These findings suggest that MMP-9 expression is more critical than MMP-2 in mediating TGF-ß-induced ASC formation.


Subject(s)
Cataract/genetics , Epithelial-Mesenchymal Transition , Lens Capsule, Crystalline/pathology , Matrix Metalloproteinase 9/genetics , Transforming Growth Factor beta1/pharmacology , Actins/metabolism , Animals , Cadherins/metabolism , Cataract/chemically induced , Matrix Metalloproteinase 2/genetics , Mice, Knockout , Mice, Transgenic
20.
Dev Dyn ; 243(10): 1298-309, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24753151

ABSTRACT

BACKGROUND: Transcription factors are critical in regulating lens development. The AP-2 family of transcription factors functions in differentiation, cell growth and apoptosis, and in lens and eye development. AP-2α, in particular, is important in early lens development, and when conditionally deleted at the placode stage defective separation of the lens vesicle from the surface ectoderm results. AP-2α's role during later stages of lens development is unknown. To address this, the MLR10-Cre transgene was used to delete AP-2α from the lens epithelium beginning at embryonic day (E) 10.5. RESULTS: The loss of AP-2α after lens vesicle separation resulted in morphological defects beginning at E18.5. By P4, a small highly vacuolated lens with a multilayered epithelium was evident in the MLR10-AP-2α mutants. Epithelial cells appeared elongated and expressed fiber cell specific ßB1 and γ-crystallins. Epithelial cell polarity and lens cell adhesion was disrupted and accompanied by the misexpression of ZO-1, N-Cadherin, and ß-catenin. Cell death was observed in the mutant lens epithelium between postnatal day (P) 14 and P30, and correlated with altered arrangements of cells within the epithelium. CONCLUSIONS: Our findings demonstrate that AP-2α continues to be required after lens vesicle separation to maintain a normal lens epithelial cell phenotype and overall lens integrity and to ensure correct fiber cell differentiation.


Subject(s)
Lens, Crystalline/physiology , Transcription Factor AP-2/physiology , Animals , Cataract/genetics , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Polarity/genetics , Embryo, Mammalian , Epithelial Cells/metabolism , Epithelial Cells/physiology , Epithelium/metabolism , Epithelium/physiology , Gene Expression Regulation, Developmental , Lens, Crystalline/embryology , Mice , Mice, Transgenic , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...