Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Biol Chem ; 289(13): 9039-52, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24554718

ABSTRACT

The accumulation of crystallin fragments in vivo and their subsequent interaction with crystallins are responsible, in part, for protein aggregation in cataracts. Transgenic mice overexpressing acylpeptide hydrolase (APH) specifically in the lens were prepared to test the role of protease in the generation and accumulation of peptides. Cataract development was seen at various postnatal days in the majority of mice expressing active APH (wt-APH). Cataract onset and severity of the cataracts correlated with the APH protein levels. Lens opacity occurred when APH protein levels were >2.6% of the total lens protein and the specific activity, assayed using Ac-Ala-p-nitroanilide substrate, was >1 unit. Transgenic mice carrying inactive APH (mt-APH) did not develop cataract. Cataract development also correlated with N-terminal cleavage of the APH to generate a 57-kDa protein, along with an increased accumulation of low molecular weight (LMW) peptides, similar to those found in aging human and cataract lenses. Nontransgenic mouse lens proteins incubated with purified wt-APH in vitro resulted in a >20% increase in LMW peptides. Crystallin modifications and cleavage were quite dramatic in transgenic mouse lenses with mature cataract. Affected lenses showed capsule rupture at the posterior pole, with expulsion of the lens nucleus and degenerating fiber cells. Our study suggests that the cleaved APH fragment might exert catalytic activity against crystallins, resulting in the accumulation of distinct LMW peptides that promote protein aggregation in lenses expressing wt-APH. The APH transgenic model we developed will enable in vivo testing of the roles of crystallin fragments in protein aggregation.


Subject(s)
Cataract/metabolism , Crystallins/metabolism , Lens, Crystalline/metabolism , Peptide Hydrolases/genetics , Amino Acid Sequence , Animals , Cataract/genetics , Cataract/pathology , Crystallins/chemistry , Gene Expression , Humans , Hydrolysis , Lens, Crystalline/pathology , Mice , Mice, Transgenic , Mutation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Hydrolases/metabolism , Promoter Regions, Genetic/genetics
2.
Biochemistry ; 53(16): 2615-23, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24697516

ABSTRACT

It has been shown that αA-mini-chaperone, a peptide representing the chaperone binding site in αA-crystallin, prevents destabilized protein aggregation. αA-Mini-chaperone has been shown to form amyloid fibrils. This study was undertaken to improve the stability of αA-mini-chaperone while preserving its anti-aggregation activity by fusing the flexible and solvent-exposed C-terminal 164-173 region of αA-crystallin to the mini-chaperone sequence DFVIFLDVKHFSPEDLT. The resulting chimeric chaperone peptide, DFVIFLDVKHFSPEDLTEEKPTSAPSS (designated CP1), was characterized. Circular dichroism studies showed that unlike αA-mini-chaperone with its ß-sheet structure, the CP1 peptide exhibited a random structure. Transmission electron microscopy (TEM) examination of the CP1 peptide incubated in a shaker at 37 °C for 72 h did not reveal amyloid fibrils, whereas αA-mini-chaperone showed distinct fibrils. Consistent with TEM observation, the thioflavin T binding assay showed an increased level of dye binding in the mini-chaperone incubated at 37 °C and subjected to shaking but not of the CP1 peptide incubated under similar conditions. The chaperone activity of the CP1 peptide was comparable to that of αA-mini-chaperone against denaturing alcohol dehydrogenase, citrate synthase, and α-lactalbumin. Transduction of both peptide chaperones to COS-7 cells showed no cytotoxic effects. The antioxidation assay involving the H2O2 treatment of COS-7 cells revealed that αA-mini-chaperone and the CP1 peptide have comparable cytoprotective properties against H2O2-induced oxidative damage in COS-7 cells. This study therefore shows that the addition of C-terminal sequence 164-173 of αA-crystallin to αA-mini-chaperone influences the conformation of αA-mini-chaperone without affecting its chaperone function or cytoprotective activity.


Subject(s)
Molecular Chaperones/metabolism , alpha-Crystallin A Chain/metabolism , Amino Acid Sequence , Animals , Benzothiazoles , COS Cells/drug effects , COS Cells/metabolism , Chlorocebus aethiops , Circular Dichroism , Hydrogen Peroxide/pharmacology , Microscopy, Electron, Transmission , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Engineering , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thiazoles/metabolism , alpha-Crystallin A Chain/chemistry , alpha-Crystallin A Chain/genetics
3.
PLoS One ; 18(4): e0284082, 2023.
Article in English | MEDLINE | ID: mdl-37079653

ABSTRACT

The cornea and cranial dura mater share sensory innervation. This link raises the possibility that pathological impulses mediated by corneal injury may be transmitted to the cranial dura, trigger dural perivascular/connective tissue nociceptor responses, and induce vascular and stromal alterations affecting dura mater blood and lymphatic vessel functionality. In this study, using a mouse model, we demonstrate for the first time that two weeks after the initial insult, alkaline injury to the cornea leads to remote pathological changes within the coronal suture area of the dura mater. Specifically, we detected significant pro-fibrotic changes in the dural stroma, as well as vascular remodeling characterized by alterations in vascular smooth muscle cell (VSMC) morphology, reduced blood vessel VSMC coverage, endothelial cell expression of the fibroblast specific protein 1, and significant increase in the number of podoplanin-positive lymphatic sprouts. Intriguingly, the deficiency of a major extracellular matrix component, small leucine-rich proteoglycan decorin, modifies both the direction and the extent of these changes. As the dura mater is the most important route for the brain metabolic clearance, these results are of clinical relevance and provide a much-needed link explaining the association between ophthalmic conditions and the development of neurodegenerative diseases.


Subject(s)
Corneal Injuries , Cranial Sutures , Humans , Skull , Connective Tissue , Dura Mater/physiology , Corneal Injuries/metabolism
4.
PLoS One ; 17(6): e0269552, 2022.
Article in English | MEDLINE | ID: mdl-35666755

ABSTRACT

Cancer cell adhesion to the endothelium is a crucial process in hematogenous metastasis, but how the integrity of the endothelial barrier and endothelial cell (EC) mechanical properties influence the adhesion between metastatic cancer cells and the endothelium remain unclear. In the present study, we have measured the adhesion between single cancer cells and two types of ECs at various growth states and their mechanical properties (elasticity) using atomic force microscopy single cell force spectroscopy. We demonstrated that the EC stiffness increased and adhesion with cancer cells decreased, as ECs grew from a single cell to a confluent state and developed cell-cell contacts, but this was reversed when confluent cells returned to a single state in a scratch assay. Our results suggest that the integrity of the endothelial barrier is an important factor in reducing the ability of the metastatic tumor cells to adhere to the vascular endothelium, extravasate and lodge in the vasculature of a distant organ where secondary metastatic tumors would develop.


Subject(s)
Adhesives , Neoplasms , Cell Adhesion , Cell Communication , Endothelial Cells , Endothelium, Vascular/metabolism , Humans , Neoplasms/metabolism
5.
Am J Physiol Cell Physiol ; 301(5): C1161-74, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21832243

ABSTRACT

Using a combination of wild-type (WT) and caveolin-2 (Cav-2) knockout along with retroviral reexpression approaches, we provide the evidence for the negative role of Cav-2 in regulating anti-proliferative function and signaling of transforming growth factor ß (TGF-ß) in endothelial cells (ECs). Although, TGF-ß had a modest inhibitory effect on WT ECs, it profoundly inhibited proliferation of Cav-2 knockout ECs. To confirm the specificity of the observed difference in response to TGF-ß, we have stably reexpressed Cav-2 in Cav-2 knockout ECs using a retroviral approach. Similar to WT ECs, the anti-proliferative effect of TGF-ß was dramatically reduced in the Cav-2 reexpressing ECs. The reduced anti-proliferative effect of TGF-ß in Cav-2-positive cells was evidenced by three independent proliferation assays: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), cell count, and bromodeoxyuridine incorporation and correlated with a loss of TGF-ß-mediated upregulation of cell cycle inhibitor p27 and subsequent reduction of the levels of hyperphosphorylated (inactive) form of the retinoblastoma protein in Cav-2 reexpressing ECs. Mechanistically, Cav-2 inhibits anti-proliferative action of TGF-ß by suppressing Alk5-Smad2/3 pathway manifested by reduced magnitude and length of TGF-ß-induced Smad2/3 phosphorylation as well as activation of activin receptor-like kinase-5 (Alk5)-Smad2/3 target genes plasminogen activator inhibitor-1 and collagen type I in Cav-2-positive ECs. Expression of Cav-2 does not appear to significantly change targeting of TGF-ß receptors I and Smad2/3 to caveolar and lipid raft microdomains as determined by sucrose fractionation gradient. Overall, the negative regulation of TGF-ß signaling and function by Cav-2 is independent of Cav-1 expression levels and is not because of changing targeting of Cav-1 protein to plasma membrane lipid raft/caveolar domains.


Subject(s)
Caveolin 2/metabolism , Cell Proliferation/drug effects , Lung/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/drug effects , Animals , Cells, Cultured , Collagen Type I/metabolism , Endothelial Cells/drug effects , Lung/drug effects , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plasminogen Activator Inhibitor 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Retinoblastoma Protein/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism
6.
Proc IAPR Int Conf Pattern Recogn ; 2020: 4317-4323, 2021 Jan.
Article in English | MEDLINE | ID: mdl-34651146

ABSTRACT

Characterizing the spatial relationship between blood vessel and lymphatic vascular structures, in the mice dura mater tissue, is useful for modeling fluid flows and changes in dynamics in various disease processes. We propose a new deep learning-based approach to fuse a set of multi-channel single-focus microscopy images within each volumetric z-stack into a single fused image that accurately captures as much of the vascular structures as possible. The red spectral channel captures small blood vessels and the green fluorescence channel images lymphatics structures in the intact dura mater attached to bone. The deep architecture Multi-Channel Fusion U-Net (MCFU-Net) combines multi-slice regression likelihood maps of thin linear structures using max pooling for each channel independently to estimate a slice-based focus selection map. We compare MCFU-Net with a widely used derivative-based multi-scale Hessian fusion method [8]. The multi-scale Hessian-based fusion produces dark-halos, non-homogeneous backgrounds and less detailed anatomical structures. Perception based no-reference image quality assessment metrics PIQUE, NIQE, and BRISQUE confirm the effectiveness of the proposed method.

7.
Am J Physiol Cell Physiol ; 298(3): C693-701, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20007452

ABSTRACT

The goal of this study was to determine whether caveolin-2 (Cav-2) is capable of controlling endothelial cell (EC) proliferation in vitro. To realize this goal, we have directly compared proliferation rates and cell cycle-associated signaling proteins between lung ECs isolated from wild-type (WT) and Cav-2 knockout (KO) mice. Using three independent proliferation assays, we have determined that Cav-2 KO ECs proliferate by ca. 2-fold faster than their WT counterparts. Cell cycle analysis by flow cytometry of propidium iodide-stained cells showed a relatively higher percentage of Cav-2 KO ECs in S and G(2)/M and lower percentage in G(o)/G(1) phases of cell cycle relative to their WT counterparts. Furthermore, an over 2-fold increase in the percentage of S phase-associated Cav-2 KO relative to WT ECs was independently determined with bromodeoxyuridine incorporation assay. Mechanistically, the increase in proliferation/cell cycle progression of Cav-2 KO ECs correlated well with elevated expression levels of predominantly S phase- and G(2)/M phase-associated cyclin A and B1, respectively. Further mechanistic analysis of molecular events controlling cell cycle progression revealed increased level of hyperphosphorylated (inactive) form of G(1) to S phase transition inhibitor, the retinoblastoma protein in hyperproliferating Cav-2 KO ECs. Conversely, the expression level of the two cyclin-dependent kinase inhibitors p16(INK4) and p27(Kip1) was reduced in Cav-2 KO ECs. Finally, increased phosphorylation (activation) of proproliferative extracellular signal-regulated kinase 1/2 was observed in hyperproliferating Cav-2 KO ECs. Overall, our data suggest that Cav-2 negatively regulates lung EC proliferation and cell cycle progression.


Subject(s)
Caveolin 2/deficiency , Cell Cycle , Cell Proliferation , Endothelial Cells/metabolism , Animals , Caveolin 2/genetics , Cells, Cultured , Cyclin A/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Flow Cytometry , Intracellular Signaling Peptides and Proteins/metabolism , Lung/blood supply , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Retinoblastoma Protein/metabolism , Signal Transduction , Time Factors
8.
Front Physiol ; 10: 1364, 2019.
Article in English | MEDLINE | ID: mdl-31736785

ABSTRACT

The contribution of cranial dura mater vascular networks, as means for maintaining brain fluid movement and balance, and as the source of significant initiators and/or contributors to neurological disorders, has been overlooked. These networks consist of both blood and lymphatic vessels. The latter were discovered recently and described as sinus-associated structures thus changing the old paradigm that central nervous system lacks lymphatics. In this study, using markers specific to blood and lymphatic endothelia, we demonstrate the existence of the complex non-sinus-associated pachymeningeal lymphatic vasculature. We further show the interrelationship and possible connections between lymphatic vessels and the dural blood circulatory system. Our novel findings reveal the presence of lymphatic-like structures that exist on their own and/or in close proximity to microvessels. Of particular interest are sub-sets of vascular complexes with dual (lymphatic and blood) vessel identity representing a unique microenvironment within the cranial dura. The close association of the systemic blood circulation and meningeal lymphatics achieved in these complexes could facilitate fluid exchange between the two compartments and constitute an alternative route for CSF drainage.

9.
Methods Mol Biol ; 1814: 515-528, 2018.
Article in English | MEDLINE | ID: mdl-29956252

ABSTRACT

Adhesive interactions between living cells or ligand-receptor interactions can be studied at the molecular level using atomic force microscopy (AFM). Adhesion force measurements are performed with functionalized AFM probes. In order to measure single ligand-receptor interactions, a cantilever with a pyramidal tip is functionalized with a bio-recognized ligand (e.g., extracellular matrix protein). The ligand-functionalized probe is then brought into contact with a cell in culture to investigate adhesion between the respective probe-bound ligand and endogenously expressed cell surface receptors (e.g., integrins or other adhesion receptor). For experiments designed to examine cell-cell adhesions, a single cell is attached to a tipless cantilever which is then brought into contact with other cultured cells. Force curves are recorded to determine the forces necessary to rupture discrete adhesions between the probe-bound ligand and receptor, or to determine total adhesion force at cell-cell contacts. Here, we describe the procedures for measuring adhesions between (a) fibronectin and α5ß1 integrin, and (b) breast cancer cells and bone marrow endothelial cells.


Subject(s)
Mechanotransduction, Cellular , Microscopy, Atomic Force/methods , Biomechanical Phenomena , Calibration , Cell Adhesion , Cell Line , Humans , Ligands , Receptors, Cell Surface/metabolism
10.
PLoS One ; 13(9): e0204418, 2018.
Article in English | MEDLINE | ID: mdl-30235349

ABSTRACT

Bone is a common site of metastasis for breast cancer and the mechanisms of metastasis are not fully elucidated. The purpose of our study was to characterize temporal and molecular dynamics of adhesive interactions between human breast cancer cells (HBCC) and human bone marrow endothelium (HBME) with piconewton resolution using atomic force microscopy (AFM). In adhesion experiments, a single breast cancer cell, MDA-MB-231 (MB231) or MDA-MB-435 (MB435) was attached to the AFM cantilever and brought into contact with a confluent HBME monolayer for different time periods (0.5 to 300 sec). The forces required to rupture individual molecular interactions and completely separate interacting cells were analyzed as measures of cell-cell adhesion. Adhesive interactions between HBME and either MB231 or MB435 cells increased progressively as cell-cell contact time was prolonged from 0.5 to 300 sec due to the time-dependent increase in the number and frequency of individual adhesive events, as well as to the involvement of stronger ligand-receptor interactions over time. Studies of the individual molecule involvement revealed that Thomsen-Friedenreich antigen (TF-Ag), galectin-3, integrin-ß1, and integrin-α3 are all contributing to HBCC/HBME adhesion to various degrees in a temporally defined fashion. In conclusion, cell-cell contact time enhances adhesion of HBCC to HBME and the adhesion is mediated, in part, by TF-Ag, galectin-3, integrin-α3, and integrin-ß1.


Subject(s)
Bone Marrow Cells/pathology , Breast Neoplasms/pathology , Cell Adhesion , Microscopy, Atomic Force , Cell Line, Tumor , Endothelium/pathology , Humans , Kinetics , Neoplasm Metastasis
11.
Mol Vis ; 13: 397-407, 2007 Mar 26.
Article in English | MEDLINE | ID: mdl-17417601

ABSTRACT

PURPOSE: Insulin and insulin-like growth factors (IGFs) are putative regulators of cell proliferation and differentiation during lens development. Transgenic mice that overexpress IGF-1 in the lens have been previously described. To further understand the ocular functions of this growth factor family, the in vivo effects of insulin expression on lens development were investigated using transgenic mice. METHODS: Expression of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in mouse lens was examined by reverse-transcriptase-polymerase chain reaction (RT-PCR) and in situ hybridization. Transgenic mice that overexpress insulin in the lens were generated using two different promoters: a fiber-cell specific alphaA-crystallin (alphaA) promoter and a modified alphaA-promoter linked to the chicken delta1-crystallin enhancer (called the deltaenalphaA promoter). The deltaenalphaA promoter is active in both lens epithelial and fiber cells. The lens phenotypes were analyzed by histology and immunohistochemistry. Protein expression was examined by western blotting. RESULTS: Normal mouse lenses express both the insulin receptor (IR) and the IGF-1 receptor (IGF-1R), and their expression is highest at the lens periphery where the germinative and transitional zones are located. In transgenic mice, insulin expression in the lens induced cataract formation. The severity of the cataracts reflected the level of transgene expression, independent of the type of promoter used. In severely affected families, the spherical shape of the lens was altered and the lenses were smaller than normal. Histological analysis showed no evidence of premature differentiation of the anterior epithelial cells. In contrast to the IGF-1 mice, insulin transgenic mice exhibited an anterior shift in the location of the germinative and transitional zones, leading to a reduction of the lens epithelial compartment. Additional alterations included expansion of the lens transitional zone, variable nuclear positioning in the lens bow region, and inhibition of fiber cell denucleation and terminal differentiation. CONCLUSIONS: Elevated intraocular insulin does not enhance proliferation nor induce differentiation of mouse lens epithelial cells. Since an increase in IGF-1 causes a posterior shift of the lens geminative and transitional zones, while an increase in insulin causes an anterior shift of these zones, our results suggest that these two growth factors may work together to control the location of this structural domain during normal lens development. Our data also suggest that increased insulin-signaling activity in the lens can antagonize the endogenous signals that are responsible for fiber cell maturation and terminal differentiation.


Subject(s)
Insulin/metabolism , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Signal Transduction , Animals , Animals, Newborn , Cataract/etiology , Cataract/metabolism , Cataract/pathology , Cell Differentiation , Cell Proliferation , Cellular Senescence , Crystallins/metabolism , Embryo, Mammalian/metabolism , In Situ Hybridization , Insulin/genetics , Lens, Crystalline/pathology , Mice , Mice, Transgenic , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
12.
Int J Dev Biol ; 48(8-9): 879-88, 2004.
Article in English | MEDLINE | ID: mdl-15558479

ABSTRACT

Growth factor signaling is implicated in the regulation of lens cell proliferation and differentiation during development. Activation of growth factor receptor tyrosine kinases is known to activate Ras proteins, small GTP-binding proteins that function as part of the signal transduction machinery. In the present study, we examined which classical Ras genes are expressed in lens cells during normal development and whether expression of an activated version of Ras is sufficient to induce either lens cell proliferation or fiber cell differentiation in transgenic mice. In situ hybridization showed H-Ras, K-Ras and N-Ras are ubiquitously expressed in all cells of the embryonic (E13.5) eye, with N-Ras showing the highest level of expression. The expression level of N-Ras decreases during later stages of embryonic development, and is nearly undetected in postnatal day 21 lenses. To generate transgenic mice, a constitutively active H-Ras mutant was linked to a chimeric regulatory element containing the mouse alphaA-crystallin promoter fused to the chick delta1-crystallin lens enhancer element. In the lenses of the transgenic mice, the transgene was expressed in both lens epithelial and fiber cells. Expression of activated Ras was sufficient to stimulate lens cell proliferation but not differentiation, implying that alternative or additional signal transduction pathways are required to induce fiber cell differentiation.


Subject(s)
Epithelial Cells/pathology , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , ras Proteins/metabolism , Animals , Antimetabolites/pharmacology , Bromodeoxyuridine/pharmacology , Cell Differentiation , Cell Proliferation , Coloring Agents/pharmacology , Enhancer Elements, Genetic , Eye Proteins/metabolism , GTP-Binding Proteins/metabolism , Hyperplasia , Immunohistochemistry , In Situ Hybridization , Intermediate Filament Proteins/metabolism , Mice , Mice, Transgenic , Models, Genetic , Mutation , Plasmids/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Promoter Regions, Genetic , Signal Transduction , Time Factors , Transgenes , delta-Crystallins/genetics , delta-Crystallins/metabolism
13.
Invest Ophthalmol Vis Sci ; 45(11): 4083-90, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15505059

ABSTRACT

PURPOSE: Both the -366/+43 and the -282/+43 mouse alphaA-crystallin (or alphaA) promoters have been effective at driving transgene expression in lens fiber cells, but not in lens epithelium. Because the chick delta1-crystallin gene is expressed in lens epithelial cells, an enhancer was borrowed from this gene and linked to the alphaA promoter. This heterogenic enhancer/promoter construct was tested in transgenic mice to see whether it was active in both lens epithelium and fiber cells while retaining lens specificity. METHODS: The third intron of the chick delta1-crystallin gene, which contains a lens enhancer element, was added to the 5' end of the mouse alphaA promoter. We refer to this chimeric regulatory element as the deltaenalphaA promoter. To test its activity, we inserted coding sequences for five different genes. Transgenic mice were generated by pronuclear microinjection. Transgene expression patterns were analyzed by either X-gal staining, in situ hybridization or immunohistochemical staining. RESULTS: When deltaenalphaA-lacZ transgenic embryos were stained with X-gal at embryonic day (E)11.5, beta-galactosidase activity was detected only in the eye. Histologic sections of the stained embryos revealed that lacZ was expressed exclusively in the lens, in both epithelial and fiber cells. Transgenic mice were also generated using either the original alphaA- or the new deltaenalphaA promoter linked to an insulin cDNA. In situ hybridizations confirmed that the short alphaA promoter targeted prenatal insulin expression specifically to the lens fiber cells, whereas the deltaenalphaA promoter was active in both lens epithelial and fiber cells. Developmental studies of the deltaenalphaA-insulin mice showed that the deltaenalphaA promoter became active at the lens pit stage and remained active in all lens cells, even at postnatal ages. The deltaenalphaA promoter also successfully directed expression of SV40 T-antigen (TAg), human E2F2, and dominant negative Sprouty2 (dn-Spry2) genes to lens epithelial and fiber cells. The lens specificity of the deltaenalphaA promoter was maintained in minigenes with different types of introns and polyadenylation signals. CONCLUSIONS: A new lens-specific regulatory element was generated-the deltaenalphaA promoter, which can drive high levels of transgene expression in both lens epithelium and fiber cells throughout development. This modified promoter can be used for future transgenic studies of signal transduction and cell cycle regulation in lens epithelial cells.


Subject(s)
Enhancer Elements, Genetic/genetics , Lens, Crystalline/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , alpha-Crystallin A Chain/genetics , delta-Crystallins/genetics , Animals , Chickens , Epithelial Cells/metabolism , Female , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation , Genes, Regulator , In Situ Hybridization , Mice , Mice, Transgenic , Transgenes
14.
Invest Ophthalmol Vis Sci ; 45(10): 3589-98, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15452066

ABSTRACT

PURPOSE: Pax6 is a transcription factor necessary for the specification and subsequent formation of the ocular lens. It is expressed in all lens cells at early stages of development. After lens formation, Pax6 expression is maintained in the lens epithelium, whereas its level abruptly decreases in differentiated fiber cells. This study is to test the hypothesis that normal fiber cell differentiation would be perturbed by sustained Pax6 expression. METHODS: Transgenic mice expressing the canonical form of mouse Pax6 were created under the control of a modified mouse alphaA-crystallin promoter. The phenotypic changes in the transgenic lens were analyzed by light and electron microscopy. The effect of ectopic Pax6 expression on the lens fiber cells was investigated by in situ hybridization, immunohistochemical staining, real-time reverse transcriptase-polymerase chain reaction (RT-PCR), and two-dimensional (2-D) gel electrophoresis. RESULTS: Transgenic mice from seven different lines all had cataracts with severity that correlated with the transgene expression level in lens fiber cells. In severely affected lines, a lumen was present between the apical surfaces of the epithelial and fiber cells, suggesting that secondary fiber cell elongation is incomplete. Electron microscopy analysis showed that the ball-and-socket interdigitations between neighboring fiber cells were underdeveloped or attenuated in the transgenic lens. Most interesting, elevated levels of Pax6 in fiber cells reduced the protein levels of transcription factor cMaf, which is known to be essential in fiber cell differentiation. Furthermore, the total amount of lens proteins was 60% less than normal in the Pax6 transgenic lens. Among the crystallins examined, the relative ratio of intact betaB1-crystallin protein to total lens protein was significantly reduced. Real-time reverse transcriptase PCR showed that the ratio of betaB1-crystallin transcript levels to total mRNA levels were reduced by 87%. CONCLUSIONS: The data demonstrate that high levels of Pax6 expression disrupt normal fiber cell differentiation and maturation.


Subject(s)
Cataract/pathology , Cell Differentiation , Eye Proteins/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Lens, Crystalline/embryology , Transcription Factors/genetics , Animals , Cataract/metabolism , Crystallins/genetics , Crystallins/metabolism , DNA-Binding Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , HMGB Proteins , Immunohistochemistry , In Situ Hybridization , Lens, Crystalline/metabolism , Lens, Crystalline/ultrastructure , Male , Mice , Mice, Transgenic , Nuclear Proteins/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-maf , RNA, Messenger/metabolism , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors
15.
Zhonghua Yan Ke Za Zhi ; 38(2): 108-11, 2002 Feb.
Article in Zh | MEDLINE | ID: mdl-11955312

ABSTRACT

OBJECTIVE: To observe the penetration of 0.5% fluconazole (FCZ) into the corneas and aqueous humors of New Zealand white rabbits following its topical application. METHODS: The rabbit corneas and aqueous humors were obtained and quantified at different times after topically applying 0.5% FCZ to the rabbit eyes. The drug levels were assayed by high performance liquid chromatography (HPLC). The pharmacokinetic parameters were calculated with nonlinear least square method by the computer. RESULTS: FCZ can rapidly penetrate into rabbit corneas. Peak corneal levels occurred essentially immediately at 2 min in the corneas (15.20 +/- 1.95) microgram/g and at 15 min after application in the aqueous (2.39 +/- 0.92 mg/L). The permeability of the corneal epithelial barrier (Kep) was 1.06 x 10(-5). The half-life (t(l/2)) of the cornea was 63.96 min, and that of the aqueous humor was 42.14 min. CONCLUSION: Topical fluconazole can be used as a topical antifungal agent to treat keratitis and needs further evaluation for treating fungal endophthalmitis.


Subject(s)
Antifungal Agents/pharmacokinetics , Aqueous Humor/metabolism , Cornea/metabolism , Epithelium, Corneal/metabolism , Fluconazole/pharmacokinetics , Animals , Antifungal Agents/therapeutic use , Female , Fluconazole/therapeutic use , Keratitis/drug therapy , Male , Models, Animal , Rabbits
16.
Cancer Res ; 74(22): 6452-62, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25269481

ABSTRACT

Caveolin-2 (Cav-2), a member of caveolin protein family, is largely different from better known caveolin-1 (Cav-1) and thus might play distinct functions. Here, we provide the first genetic evidence suggesting that host-expressed Cav-2 promotes subcutaneous tumor growth and tumor-induced neovascularization using two independent syngeneic mouse models. Host deficiency in Cav-2 resulted in defective and reduced growth of subcutaneously implanted Lewis lung carcinoma (LLC) and B16-F10 melanoma tumors, respectively. Consistent with the defective growth, LLC and B16-F10 melanoma tumors implanted into Cav-2 KO mice displayed reduced microvascular density (MVD) determined by IHC with anti-CD31 antibodies, suggesting impaired pathologic angiogenesis. Additional studies involving LLC tumors extracted from Cav-2 KO mice just 10 days after implantation determined reduced cell proliferation, massive necrotic cell death, and fibrosis. In contrast with day 10, only MVD but not cell proliferation and survival was reduced in the earliest palpable LLC tumors extracted 6 days after implantation into Cav-2 KO mice, suggesting that impaired angiogenesis is the causative factor. Mechanistically, impaired LLC tumor growth and angiogenesis in Cav-2 KO mice was associated with increased expression levels of antiangiogenic thrombospondin-1 and inhibited S1177 phosphorylation of endothelial nitric oxide synthase. Taken together, our data suggest that host deficiency in Cav-2 impairs tumor-induced angiogenesis, leading to compromised tumor cell survival/proliferation manifested by the defective tumor growth. In conclusion, host-expressed Cav-2 may promote tumor growth via supporting tumor-induced angiogenesis. Thus, Cav-2 expressed in tumor microenvironment may potentially become a novel target for cancer therapy.


Subject(s)
Carcinoma, Lewis Lung/pathology , Caveolin 2/physiology , Neovascularization, Pathologic/prevention & control , Animals , Carcinoma, Lewis Lung/blood supply , Cell Proliferation , Fibrosis , Ki-67 Antigen/analysis , Male , Mice , Mice, Inbred C57BL , Necrosis , Nitric Oxide Synthase Type III/metabolism , Thrombospondin 1/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology
17.
Invest Ophthalmol Vis Sci ; 55(8): 4731-40, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24994865

ABSTRACT

PURPOSE: Posterior capsule opacification (PCO) after cataract surgery is due in part to proliferation of the adhering lens epithelial cells and transdifferentiation into mesenchymal cells. The histone deacetylase (HDAC) inhibitors, trichostatin A (TSA) and vorinostat (suberoylanilidehydroxamic acid [SAHA]) are known to modulate cell proliferation and epithelial-mesenchymal transition (EMT). Studies have shown that TGFß2 can induce EMT similar to that seen during PCO. This study evaluated the effects of TSA and SAHA on TGFß2-induced EMT in lens epithelial explants. METHODS: Epithelial cells adherent to lens capsules were isolated from fresh pig lenses and human donor lenses and cultured for 12 hours. Explants were pretreated with TSA or SAHA for 1 hour and then treated with TGFß2 for up to 3 days. Scratch wound healing assay was used to determine epithelial cell proliferation and migration in the samples. The effects of TSA and SAHA on histone acetylation and HDAC 1 to 6 levels were analyzed by Western blotting. RESULTS: Western blotting and immunocytochemistry demonstrated high expression of α-SMA in lens epithelial cells treated with TGFß2. The HDAC inhibitors exerted dose-dependent inhibition of α-SMA expression, with complete inhibition occurring with 0.5 µM of TSA and 2.5 µM of SAHA. Transforming growth factor ß2-induced EMT was suppressed by TSA and SAHA. Histone deacetylase inhibition in pig lens epithelia led to increased acetylation of histone 3 and 4 at multiple sites. CONCLUSIONS: Histone deacetylase inhibitors, TSA, and SAHA prevent EMT in lens epithelial explants. The results also suggest that the epigenetic modifiers are the potential targets to control PCO after cataract surgery.


Subject(s)
Actins/biosynthesis , Capsule Opacification/prevention & control , Epithelial Cells/metabolism , Hydroxamic Acids/pharmacology , Lens Capsule, Crystalline/metabolism , Transforming Growth Factor beta2/adverse effects , Actins/drug effects , Animals , Blotting, Western , Capsule Opacification/etiology , Capsule Opacification/metabolism , Cataract Extraction/adverse effects , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Fluorine Radioisotopes , Histone Deacetylase Inhibitors/pharmacology , Humans , Immunohistochemistry , Lens Capsule, Crystalline/drug effects , Lens Capsule, Crystalline/pathology , Microscopy, Fluorescence , Middle Aged , Swine , Transforming Growth Factor beta2/metabolism , Vorinostat
18.
FEBS Lett ; 586(19): 3317-23, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22819829

ABSTRACT

Here we show that tyrosine phosphorylation of caveolin-2 (Cav-2) negatively regulates the anti-proliferative function of transforming growth factor beta (TGF-beta) in endothelial cells. In contrast to wild-type-Cav-2, retroviral re-expression of Y19/27F-Cav-2 in Cav-2 knockout endothelial cells did not affect anti-proliferative effect of TGF-beta compared to empty vector. Conversely, although less effective than wild-type, re-expression of S23/36A-Cav-2 reduced the effect of TGF-beta compared to empty vector. This differential effect of tyrosine and serine phosphorylation mutants of Cav-2 correlated with TGF-beta-induced Smad3 phosphorylation and transcriptional activation of plasminogen activator inhibitor-1. Thus tyrosine-phosphorylated Cav-2 counteracts anti-proliferative effect of TGF-beta in endothelial cells.


Subject(s)
Caveolin 2/chemistry , Caveolin 2/metabolism , Transforming Growth Factor beta/pharmacology , Amino Acid Substitution , Animals , Caveolin 2/antagonists & inhibitors , Caveolin 2/genetics , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Knockout Techniques , Humans , Mice , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphorylation , Plasminogen Activator Inhibitor 1/genetics , Protein Tyrosine Phosphatases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Serpin E2/genetics , Smad3 Protein/metabolism , Transcriptional Activation/drug effects , Tyrosine/chemistry
19.
Brain Res Bull ; 81(2-3): 287-96, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19897021

ABSTRACT

PURPOSE: Transforming growth factor beta (TGFbeta) is an important cytokine in corneal development and wound healing. Transgenic mice that express an active form of human TGFbeta1 driven by a lens-specific promoter were used in the current study to determine the biological effects of lens-derived TGFbeta1 on postnatal corneal development and homeostasis. METHODS: The postnatal corneal changes in the TGFbeta1 transgenic mice were examined by fluorescein labeling and histology. Epithelial/endothelial-to-mesenchymal transition (E/EnMT) in the transgenic mouse cornea was demonstrated by immunostaining for alpha-smooth muscle actin (alpha-SMA) and cadherin-11. Expression of E- and N-cadherin in the corneal epithelial and endothelial cells, respectively, was analyzed by in situ hybridization. RESULTS: Among the established TGFbeta1 transgenic lines, mice from line OVE853 and OVE917 had normal-sized eyeballs but developed a corneal haze after eyelid opening. Histological examination showed that prenatal corneal development appeared to be normal. However, after postnatal day 7 (P7), the corneal endothelial cells in transgenic line OVE853 began to lose normal cell-cell contact and basement membrane structure. The endothelial layer was eventually absent in the inner surface of the transgenic mouse cornea. The morphological changes in the cornea correlated with abnormal expression of alpha-SMA, a molecular marker of EMT, and stress fiber formation in myofibroblast-like cells, which initially appeared in the corneal endothelial layer and subsequently in the corneal epithelial and stromal layers. The E/EnMT in the transgenic mouse cornea was further demonstrated by loss of E- and N-cadherin expression in the corneal epithelial and endothelial cells, respectively, and meanwhile increasing expression of cadherin-11 in both corneal epithelium and stroma. CONCLUSIONS: Elevated levels of active TGFbeta1 in the anterior chamber can lead to myofibroblast formation in the corneal endothelial layer and subsequently in the corneal epithelial and stromal layers. Our data suggest that the levels of biologically active TGFbeta in the aqueous humor must be under tight control to maintain corneal homeostasis. TGFbeta1 is the major cytokine during wound healing. Therefore, our findings also suggest a potential mechanism to explain the loss of corneal endothelial barrier and corneal opacification after intraocular surgery or trauma.


Subject(s)
Corneal Stroma/cytology , Fibroblasts/physiology , Gene Expression Regulation, Developmental/physiology , Transforming Growth Factor beta1/pharmacology , Actins/metabolism , Animals , Animals, Newborn , Cadherins/metabolism , Cell Differentiation/drug effects , Corneal Stroma/growth & development , Fibroblasts/drug effects , Humans , Mice , Mice, Transgenic , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics
20.
Biochemistry ; 47(1): 101-11, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18081315

ABSTRACT

In the present study, using a combination of reconstituted systems and endothelial cells endogenously expressing caveolins, we show that phosphorylation of caveolin-2 at serines 23 and 36 can be differentially regulated by caveolin-1 mediated subcellular targeting to lipid raft/caveolae and in endothelial cells synchronized in mitosis. Detergent insolubility and sucrose flotation gradient experiments revealed that serine 23 phosphorylation of caveolin-2 preferably occurs in detergent-resistant membranes (DRMs), while serine 36 phosphorylation takes place in non-DRMs. Furthermore, immunofluorescence microscopy studies determined that in the presence of caveolin-1, serine 23-phosphorylated caveolin-2 mostly localizes to plasma membrane, while serine 36-phosphorylated caveolin-2 primarily resides in intracellular compartments. To directly address the role of caveolin-1 in regulating phosphorylation of endogenous caveolin-2, we have used the siRNA approach. The specific knockdown of caveolin-1 in endothelial cells decreases caveolin-2 phosphorylation at serine 23 but not at serine 36. Thus, upregulation of serine 23 phosphorylation of caveolin-2 depends on caveolin-1-driven targeting to plasma membrane lipid rafts and caveolae. Interestingly, although serine 36 phosphorylation does not seem to be regulated in endothelial cells by caveolin-1, it can be selectively upregulated in endothelial cells synchronized in mitosis. The latter data suggests a possible involvement of serine 36-phosphorylated caveolin-2 in modulating mitosis.


Subject(s)
Caveolae/metabolism , Caveolin 2/metabolism , Endothelial Cells/metabolism , Membrane Microdomains/metabolism , Serine/metabolism , Caveolin 1/chemistry , Caveolin 1/genetics , Caveolin 1/metabolism , Caveolin 2/chemistry , Caveolin 2/genetics , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Detergents/chemistry , Dimerization , Endothelial Cells/cytology , Flow Cytometry , Humans , Microscopy, Fluorescence , Mitosis/physiology , Phosphorylation , Protein Transport , RNA, Small Interfering/genetics , Serine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL