Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
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.
Mol Brain ; 16(1): 83, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38124148

ABSTRACT

Unraveling the intricate relationship between mechanical factors and brain activity is a pivotal endeavor, yet the underlying mechanistic model of signaling pathways in brain mechanotransduction remains enigmatic. To bridge this gap, we introduced an in situ multi-scale platform, through which we delineate comprehensive brain biomechanical traits in white matter (WM), grey-white matter junctions (GW junction), and the pons across human brain tissue from four distinct donors. We investigate the three-dimensional expression patterns of Piezo1, Piezo2, and TMEM150C, while also examining their associated histological features and mechanotransduction signaling networks, particularly focusing on the YAP/ß-catenin axis. Our results showed that the biomechanical characteristics (including stiffness, spring term, and equilibrium stress) associated with Piezo1 vary depending on the specific region. Moving beyond Piezo1, our result demonstrated the significant positive correlations between Piezo2 expression and stiffness in the WM. Meanwhile, the expression of Piezo2 and TMEM150C was shown to be correlated to viscoelastic properties in the pons and WM. Given the heterogeneity of brain tissue, we investigated the three-dimensional expression of Piezo1, Piezo2, and TMEM150C. Our results suggested that three mechanosensitive proteins remained consistent across different vertical planes within the tissue sections. Our findings not only establish Piezo1, Piezo2, and TMEM150C as pivotal mechanosensors that regulate the region-specific mechanotransduction activities but also unveil the paradigm connecting brain mechanical properties and mechanotransduction activities and the variations between individuals.


Subject(s)
Ion Channels , Mechanotransduction, Cellular , Humans , Brain/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology
3.
Cornea ; 41(10): 1325-1336, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36107852

ABSTRACT

ABSTRACT: The cornea is subject to a myriad of ocular conditions often attributed to cell loss or cell dysfunction. Owing to the superficial positioning of tissues composing the anterior segment of the eye, particularly the cornea, regenerative medicine in this region is aided by accessibility as compared with the invasive delivery methods required to reach deep ocular tissues. As such, cell therapies employing the use of carrier substrates have been widely explored. This review covers recent advances made in the delivery of stem cells, corneal epithelial cells, and corneal endothelial cells. Particular focus is placed on the most popular forms of synthetic scaffolds currently being examined: contact lenses, electrospun substrates, polymeric films, and hydrogels.


Subject(s)
Biocompatible Materials , Contact Lenses , Cornea , Endothelial Cells , Humans , Hydrogels
4.
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.

5.
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
6.
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
7.
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
8.
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
9.
Biotechnol Bioeng ; 118(1): 423-432, 2021 01.
Article in English | MEDLINE | ID: mdl-32970320

ABSTRACT

Vascular leak is a key driver of organ injury in diseases, and strategies that reduce enhanced permeability and vascular inflammation are promising therapeutic targets. Activation of the angiopoietin-1 (ANG1)-Tie2 tyrosine kinase signaling pathway is an important regulator of vascular quiescence. Here we describe the design and construction of a new soluble ANG1 mimetic that is a potent activator of endothelial Tie2 in vitro and in vivo. Using a chimeric fusion strategy, we replaced the extracellular matrix (ECM) binding and oligomerization domain of ANG1 with a heptameric scaffold derived from the C-terminus of serum complement protein C4-binding protein α. We refer to this new fusion protein biologic as Hepta-ANG1, which forms a stable heptamer and induces Tie2 phosphorylation in cultured cells, and in the lung following intravenous injection of mice. Injection of Hepta-ANG1 ameliorates vascular endothelial growth factor- and lipopolysaccharide-induced vascular leakage, in keeping with the known functions of Angpt1-Tie2 in maintaining quiescent vascular stability. The new Hepta-ANG1 fusion is easy to produce and displays remarkable stability with high multimericity that can potently activate Tie2. It could be a new candidate ANG1 mimetic therapy for treatments of inflammatory vascular leak, such as acute respiratory distress syndrome and sepsis.


Subject(s)
Angiopoietin-1 , Complement C4b-Binding Protein , Human Umbilical Vein Endothelial Cells/metabolism , Recombinant Fusion Proteins , Vascular Diseases/drug therapy , Angiopoietin-1/biosynthesis , Angiopoietin-1/genetics , Angiopoietin-1/pharmacology , Animals , Complement C4b-Binding Protein/biosynthesis , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/pharmacology , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Protein Domains , Rabbits , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/pathology , Vascular Diseases/metabolism , Vascular Diseases/pathology
10.
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
11.
Cell Mol Life Sci ; 77(14): 2701-2722, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32008085

ABSTRACT

Epithelial to mesenchymal transition (EMT) is a complex plastic and reversible cellular process that has critical roles in diverse physiological and pathological phenomena. EMT is involved in embryonic development, organogenesis and tissue repair, as well as in fibrosis, cancer metastasis and drug resistance. In recent years, the ability to edit the genome using the clustered regularly interspaced palindromic repeats (CRISPR) and associated protein (Cas) system has greatly contributed to identify or validate critical genes in pathway signaling. This review delineates the complex EMT networks and discusses recent studies that have used CRISPR/Cas technology to further advance our understanding of the EMT process.


Subject(s)
CRISPR-Cas Systems/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Editing/methods , Embryonic Development/genetics , Humans , Organogenesis/genetics , Signal Transduction/genetics
12.
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
13.
Dis Model Mech ; 11(6)2018 06 15.
Article in English | MEDLINE | ID: mdl-29752281

ABSTRACT

The bones of the cranial vault are formed directly from mesenchymal cells through intramembranous ossification rather than via a cartilage intermediate. Formation and growth of the skull bones involves the interaction of multiple cell-cell signaling pathways, with fibroblast growth factors (FGFs) and their receptors exerting a prominent influence. Mutations within the FGF signaling pathway are the most frequent cause of craniosynostosis, which is a common human craniofacial developmental abnormality characterized by the premature fusion of the cranial sutures. Here, we have developed new mouse models to investigate how different levels of increased FGF signaling can affect the formation of the calvarial bones and associated sutures. Whereas moderate Fgf8 overexpression resulted in delayed ossification followed by craniosynostosis of the coronal suture, higher Fgf8 levels promoted a loss of ossification and favored cartilage over bone formation across the skull. By contrast, endochondral bones were still able to form and ossify in the presence of increased levels of Fgf8, although the growth and mineralization of these bones were affected to varying extents. Expression analysis demonstrated that abnormal skull chondrogenesis was accompanied by changes in the genes required for Wnt signaling. Moreover, further analysis indicated that the pathology was associated with decreased Wnt signaling, as the reduction in ossification could be partially rescued by halving Axin2 gene dosage. Taken together, these findings indicate that mesenchymal cells of the skull are not fated to form bone, but can be forced into a chondrogenic fate through the manipulation of FGF8 signaling. These results have implications for evolution of the different methods of ossification as well as for therapeutic intervention in craniosynostosis.


Subject(s)
Chondrogenesis , Fibroblast Growth Factor 8/metabolism , Osteogenesis , Signal Transduction , Skull/embryology , Skull/metabolism , Alleles , Animals , Axin Protein/genetics , Bone and Bones/pathology , Cartilage/abnormalities , Cartilage/pathology , Cell Differentiation/genetics , Chondrogenesis/genetics , Craniosynostoses/genetics , Craniosynostoses/pathology , Fibroblast Growth Factor 8/genetics , Gene Dosage , Gene Expression Regulation, Developmental , Mice , Mutation/genetics , Osteogenesis/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Wnt Signaling Pathway/genetics
14.
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
15.
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.

16.
Biochim Biophys Acta ; 1813(1): 213-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20883729

ABSTRACT

HSP90, a major molecular chaperone, plays an essential role in the maintenance of several signaling molecules. Inhibition of HSP90 by inhibitors such as 17-allylamino-demethoxy-geldanamycin (17AAG) is known to induce apoptosis in various cancer cells by decreasing the activation or expression of pro-survival molecules such as protein kinase B (Akt). While we did not observe either decrease in expression or activation of pro-survival signaling molecules in human breast cancer cells upon inhibiting HSP90 with 17AAG, we did observe a decrease in cell motility of transformed cells, and cell motility and invasion of cancer cells. We found a significant decrease in the number of filopodia and lamellipodia, and in the F-actin bundles upon HSP90 inhibition. Our results show no change in the active forms or total levels of FAK and Pax, or in the activation of Rac-1 and Cdc-42; however increased levels of HSP90, HSP90α and HSP70 were observed upon HSP90 inhibition. Co-immuno-precipitation of HSP90 reveals interaction of HSP90 with G-actin, which increases upon HSP90 inhibition. FRET results show a significant decrease in interaction between actin monomers, leading to decreased actin polymerization upon HSP90 inhibition. We observed a decrease in the invasion of human breast cancer cells in the matrigel assay upon HSP90 inhibition. Over-expression of αB-crystallin, known to be involved in actin dynamics, did not abrogate the effect of HSP90 inhibition. Our work provides the molecular mechanism by which HSP90 inhibition delays cell migration and should be useful in developing cancer treatment strategies with known anti-cancer drugs such as cisplatin in combination with HSP90 inhibitors.


Subject(s)
Actins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Benzoquinones/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Cell Line, Tumor , Collagen/metabolism , Drug Combinations , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lactams, Macrocyclic/pharmacology , Laminin/metabolism , Proteoglycans/metabolism , Signal Transduction , Wound Healing
17.
Biochem Pharmacol ; 78(2): 142-52, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19464431

ABSTRACT

Heat shock protein 90 (Hsp90) is a major molecular chaperone that plays an essential role in the maintenance of several signaling molecules, most of which are oncogenic kinases. Hsp90 inhibition by specific inhibitors leads to destabilization and loss of activity of such proteins, thereby leading to inhibition of multiple signaling cascades. Due to this, Hsp90 has emerged as an important target for the treatment of cancer. Inhibition of Hsp90 has been reported to induce apoptosis in certain cancer cell types. However, the molecular details of induction of apoptosis upon Hsp90 inhibition are not understood. We have investigated the effect of Hsp90 inhibition on a non-adherent rat histiocytoma cell line, BC-8, using geldanamycin and 17-Allylamino-17-demethoxygeldanamycin. We show that Hsp90 inhibition induces ER stress, which leads to disruption of mitochondrial homeostasis, leading to apoptosis. Induction of ER stress leads to increased expression of ER chaperones, Grp78 and Grp94, cleavage of caspase-12 and increase in cytoplasmic calcium. We show that calcium and Bax are responsible for the decrease in mitochondrial membrane potential (Deltapsi(m)), thereby leading to the release of cytochrome c and activation of caspase-9. Moreover, calcium chelator and over-expression of Bcl-2 is able to confer protection against apoptosis upon Hsp90 inhibition. We conclude that inhibition of Hsp90 leads to ER stress-induced mitochondria-mediated apoptosis and that Bax and Ca(2+) play an important role in mitochondrial damage.


Subject(s)
Apoptosis/physiology , Benzoquinones/pharmacology , Endoplasmic Reticulum/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Histiocytoma/metabolism , Histiocytoma/pathology , Lactams, Macrocyclic/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , HSP90 Heat-Shock Proteins/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Rats , Stress, Physiological/drug effects , Stress, Physiological/physiology
18.
J Mol Biol ; 382(3): 812-24, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18692065

ABSTRACT

Oxidative stress and Cu(2+) have been implicated in several neurodegenerative diseases and in cataract. Oxidative stress, as well as Cu(2+), is also known to induce the expression of the small heat shock proteins alpha-crystallins. However, the role of alpha-crystallins in oxidative stress and in Cu(2+)-mediated processes is not clearly understood. We demonstrate using fluorescence and isothermal titration calorimetry that alpha-crystallins (alphaA- and alphaB-crystallin and its phosphorylation mimic, 3DalphaB-crystallin) bind Cu(2+) with close to picomolar range affinity. The presence of other tested divalent cations such as Zn(2+), Mg(2+), and Ca(2+) does not affect Cu(2+) binding, indicating selectivity of the Cu(2+)-binding site(s) in alpha-crystallins. Cu(2+) binding induces structural changes and increase in the hydrodynamic radii of alpha-crystallins. Cu(2+) binding increases the stability of alpha-crystallins towards guanidinium chloride-induced unfolding. Chaperone activity of alphaA-crystallin increases significantly upon Cu(2+) binding. Alpha-crystallins rescue amyloid beta peptide, Abeta(1-40), from Cu(2+)-induced aggregation in vitro. Alpha-crystallins inhibit Cu(2+)-induced oxidation of ascorbate and, hence, prevent the generation of reactive oxygen species. Interestingly, alpha-synuclein, a Cu(2+)-binding protein, does not inhibit this oxidation process significantly. We find that the Cu(2+)-sequestering (or redox-silencing) property of alpha-crystallins confers cytoprotection. To the best of our knowledge, this is the first study to reveal high affinity (close to picomolar) for Cu(2+) binding and redox silencing of Cu(2+) by any heat shock protein. Thus, our study ascribes a novel functional role to alpha-crystallins in Cu(2+) homeostasis and helps in understanding their protective role in neurodegenerative diseases and cataract.


Subject(s)
Copper/metabolism , Heat-Shock Proteins, Small/metabolism , Oxidation-Reduction , alpha-Crystallin A Chain/metabolism , alpha-Crystallin B Chain/metabolism , Antioxidants/metabolism , Ascorbic Acid/metabolism , Binding Sites , Calorimetry , Cell Line , Copper/chemistry , Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/genetics , Humans , Oxidative Stress , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism , Spectrometry, Fluorescence , alpha-Crystallin A Chain/chemistry , alpha-Crystallin A Chain/genetics , alpha-Crystallin B Chain/chemistry , alpha-Crystallin B Chain/genetics
19.
J Biosci ; 32(3): 585-93, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17536178

ABSTRACT

Heat shock response is associated with the synthesis of heat shock proteins (Hsps) which is strictly regulated by different members of heat shock transcription factors (HSFs). We previously reported that a rat histiocytoma, BC-8 failed to synthesize Hsps when subjected to typical heat shock conditions (42 degrees C, 60 min). The lack of Hsp synthesis in these cells was due to a failure in HSF1 DNA binding activity. In the present study we report that BC-8 tumor cells when subjected to heat shock at higher temperature (43 degrees C, 60 min) or incubation for longer time at 42 degrees C, exhibited necrosis characteristics; however,under mild heat shock (42 degrees C, 30 min) conditions cells showed activation of autophagy. Mild heat shock treatment induced proteolysis of HSF1, and under similar conditions we observed an increase in HSF2 expression followed by its enhanced DNA binding activity. Inhibiting HSF1 proteolysis by reversible proteasome inhibition failed to inhibit heat shock induced autophagy. Compromising HSF2 expression but not HSF1 resulted in the inhibition of autophagy, suggesting HSF2 dependent activation of autophagy. We are reporting for the first time that HSF2 is heat inducible and functions in heat shock induced autophagic cell death in BC-8 tumor cells.


Subject(s)
Cell Death/physiology , DNA-Binding Proteins/metabolism , Heat-Shock Proteins/metabolism , Histiocytoma/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Gene Expression Regulation , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Hot Temperature , Rats , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...